Next Article in Journal
Examination of Anti-Inflammatory Effects After Propionate Supplementation in the R6/2 Mouse Model of Huntington’s Disease
Previous Article in Journal
Deciphering the Role of Rhodanine Flanked Non-Fullerene Acceptor Molecules for Efficient Organic Photovoltaics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intracranial Aneurysm Biomarkers: A Convergence of Genetics, Inflammation, Oxidative Stress, and the Extracellular Matrix

1
Ningbo Key Laboratory of Nervous System and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
2
Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(7), 3316; https://doi.org/10.3390/ijms26073316
Submission received: 13 March 2025 / Revised: 30 March 2025 / Accepted: 30 March 2025 / Published: 2 April 2025
(This article belongs to the Section Molecular Neurobiology)

Abstract

Intracranial aneurysm (IA) is a common cerebrovascular disease in which sacral aneurysms occurring in the Wills ring region can lead to devastating subarachnoid hemorrhage. Despite advances in research, the underlying mechanisms of IA formation and rupture remain incompletely understood, hindering early diagnosis and effective treatment. This review comprehensively summarizes the current landscape of IA biomarkers, encompassing genetic markers, DNA, RNA, inflammatory molecules, oxidative stress proteins, and extracellular matrix (ECM) components. Accumulating evidence suggests that various biomarkers are associated with different stages of IA pathogenesis, including initiation, progression, and rupture. Aberrant ECM composition and remodeling have been observed in IA patients, and extracellular matrix-degrading enzymes are implicated in IA growth and rupture. Biomarker research in IA holds great potential for improving clinical outcomes. Future studies should focus on validating the existing biomarkers, identifying novel ones, and investigating their underlying mechanisms to facilitate the development of personalized preventive and therapeutic strategies for IA.

1. Background

Intracranial aneurysm (IA), a prevalent cerebrovascular disease, occurs in 1–2% of the population [1]. It mainly occurs in the arterial system at the bottom of the brain, especially within the Willis ring, also known as sacral (Berry) aneurysm. The hallmark feature of IA is localized alteration of the cerebral artery wall structure, including the absence of the internal elastic lamina (IEL) and media rupture (Figure 1) [2]. In a cross-sectional study conducted in China, 7% of adults aged 35 to 75 were found to have aneurysms through extensive screening using magnetic resonance angiography (MRA) [3]. In a prevalence study based on a European population, approximately 1.8% of adult participants were found to have aneurysms through screening MRI [4]. Unruptured intracranial aneurysms are more common in women than in men, with a female-to-male ratio of 3:1 in large population studies [5]. They are also more prevalent in the elderly and are rare in children [6]. Untreated IA can rupture any time, with subarachnoid hemorrhage (SAH) being a frequent consequence. Among SAH patients, approximately 12% die before reaching medical attention, 40% die within one month of onset, and over 1/3 of survivors suffer permanent neurological deficits [7].
Due to the high mortality and disability associated with IA rupture, early identification and treatment are crucial. Early IA is often asymptomatic, necessitating early screening for diagnosis. Angiography is a common tool for early detection and diagnosis. Digital subtraction angiography (DSA) remains the gold standard for IA diagnosis; however, it is invasive and time-consuming. Non-invasive techniques like computed tomography angiography (CTA) are increasingly used. While CTA offers increased accessibility, it may lack sensitivity for smaller aneurysms, and it requires radiation exposure and contrast agents, which can be problematic for patients with renal dysfunction or contrast agent allergies [14]. Magnetic resonance angiography (MRA) is another non-invasive method with diagnostic accuracy similar to CTA, and it avoids the aforementioned issues with contrast agents. However, MRA has limitations in specificity [15]. The absence of readily available non-invasive biomarkers for IA diagnosis underscores the need for discovering novel markers to predict IA presence and rupture risk.

2. The Risk Factors and Causes of Aneurysm Formation and Rupture

Intracranial aneurysms (IAs) are characterized by localized changes in the structure of the cerebral artery walls. These changes include the absence of the IEL and media rupture. The etiology of IA formation and rupture is complex and not fully understood, but it likely involves a combination of genetic susceptibility, modifiable vascular risk factors (including hypertension, lipid accumulation, atherosclerosis, and smoking), and hemodynamic stress [8].
Healthy cerebral arteries consist of three distinct layers: the intima (innermost), media, and adventitia (outermost) [16]. The IEL, a crucial structure within the media, contributes to IA formation [16]. Compared with other arteries, intracranial arteries have a thicker IEL with a lower proportion of elastic fibers and smooth muscle cells (SMCs) in the media and a thinner adventitia with sparser connective tissue in the subarachnoid space. These anatomical features may render them more susceptible to aneurysm formation [16].
Genetic susceptibility plays a role in IA, with a large-scale meta-analysis identifying 19 single nucleotide polymorphisms (SNPs) associated with sporadic IA [17]. However, the largest twin study to date did not find significant genetic contributions, suggesting a complex interplay between genes and environment [18]. Modifiable risk factors, such as smoking and hypertension, appear to have a more decisive role. A case–control study reported that smoking and hypertension are strong risk factors for IA [19].
Hemodynamic stress is another important factor. IA commonly occurs at arterial bifurcations, branching points, or locations with abrupt changes in vascular geometry, where excessive hemodynamic stress is exerted on the arterial wall [20]. While some studies suggest high wall shear stress (WSS) and WSS gradients contribute to aneurysm formation [20], others suggest low WSS may promote the growth and rupture of large atherosclerotic aneurysms, while high WSS may drive the development and rupture of small aneurysms [21]. This highlights the complex role of hemodynamic forces in IA pathogenesis.
The common steps in IA formation include endothelial dysfunction/injury, exacerbated inflammatory responses, vascular smooth muscle cell (VSMC) phenotype modulation, extracellular matrix remodeling, followed by subsequent cell death and vascular wall degeneration [8]. Risk factors can lead to the rupture of the IEL, causing abnormal blood flow and mechanical stress on the arterial wall. This, in turn, triggers SMC apoptosis or modulation, endothelial dysfunction, and macrophage influx, further promoting inflammatory responses and extracellular matrix degradation [8]. Macrophages play a crucial role in the inflammatory response, with two distinct populations having opposing effects. Unruptured IA exhibits a balance between pro-inflammatory M1 and anti-inflammatory M2 macrophages, while ruptured IA shows an increase in M1 cells, suggesting a role for M1/M2 imbalance in rupture [22]. Early IA formation involves SMC migration to the intima and proliferation due to endothelial injury, leading to intimal hyperplasia. Subsequently, SMCs undergo a phenotypic switch from contractile to dedifferentiated, promoting inflammation and matrix degradation [23]. The inflammatory response further damages the arterial wall through the activation of molecules like tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin-1β (IL-1β), Nuclear factor kappa-B (NF-κB), matrix metalloproteinases (MMPs), cyclooxygenase-1 (COX-1), and cyclooxygenase-2 (COX-2) [8]. The influx of blood exposes structurally weak arteries to high wall shear forces, leading to the formation of aneurysmal sacs. The aneurysmal sac grows until a balance is reached between ongoing vascular wall repair and extracellular matrix degradation [12] (Figure 1). The precise molecular mechanisms and inflammatory mediators leading to aneurysm rupture remain uncertain.

3. Research Progress on Biomarkers for Intracranial Aneurysms

Biomarkers are molecules that can indicate the presence or severity of a disease. In recent years, the development of “omics” technologies (genomics, transcriptomics, proteomics, and metabolomics) has accelerated the discovery of potential biomarkers for IA [24]. These biomarkers include nucleic acids, proteins, sugars, lipids, small metabolites, and extracellular vesicles (exosomes). For example, miR-29a was identified as a potential biomarker for early detection of IA and a prognostic indicator for the course of the disease by Wang et al. [25]. Similarly, Kao et al. suggested that plasma levels of interleukin-6 (IL-6) may be an early predictor of outcomes in patients with ruptured IA [26].
This review summarizes the potential biomarkers for IA identified in recent years, including extracellular vesicles, circulating nucleic acids (DNA, RNA, and microRNA [miRNA]), and proteins (Figure 2). There is considerable promise in these biomarkers for the early detection of IA, allowing for interventions before aneurysm rupture. Early diagnosis can significantly improve treatment success rates, reduce mortality rates, and enhance patient quality of life.

4. Sources of Biomarkers for IA

Biomarkers for IA can be categorized based on their source into tissue-derived, blood-derived, and cerebrospinal fluid (CSF)-derived biomarkers (Figure 2). Tissue biomarkers are primarily derived from affected organs or pathological sites and are obtained through biopsy, surgical resection, or autopsy. These biomarkers typically include specific gene mutations, changes in RNA expression levels, proteomic characteristics, and alterations in the tissue microenvironment [30]. In IA research, tissue biomarkers can be used to analyze structural changes in the vascular wall, the expression of inflammatory factors, and the characteristics of extracellular matrix (ECM) remodeling [33]. However, the acquisition of tissue samples is relatively invasive, which limits their clinical application. Blood is one of the most important sources of biomarkers, offering advantages such as ease of collection, repeatability of detection, and strong potential for clinical translation. Blood biomarkers primarily include circulating RNAs (such as miRNA and lncRNA), cell-free DNA (cfDNA), proteins (including inflammatory factors, enzymes, and receptors), and extracellular vesicles (EVs). In intracranial aneurysm research, blood biomarkers are widely used to assess inflammation status, oxidative stress levels, and changes in gene expression [31]. Additionally, due to the regulatory role of the blood–brain barrier, changes in specific biomarkers in the blood may indirectly reflect the progression of brain diseases [34]. CSF is directly connected to the central nervous system (CNS) and serves as a crucial carrier of biomarkers for neurological diseases [35]. CSF biomarkers include proteins (such as neuroinjury-related proteins), non-coding RNAs, metabolic products, and extracellular vesicles. Since CSF more directly reflects the physiological and pathological changes in intracranial lesions compared with blood, it exhibits higher sensitivity in cerebrovascular disease research [32]. In patients with intracranial aneurysms, inflammatory factors, extracellular matrix degradation products, and neuron injury-related molecules in CSF may serve as important diagnostic and prognostic biomarkers [36].

5. DNA

Genetic variations and epigenetic abnormalities in DNA significantly impact IA formation and rupture. Roder et al. analyzed gene expression data from IA tissue samples, identifying seven genes (B-cell lymphoma-2 (BCL2), collagen type I alpha 2 chain (COL1A2), collagen type III alpha 1 chain (COL3A1), collagen type V alpha 2 chain (COL5A2), chemokine (C-X-C motif) ligand 12 (CXCL12), tissue inhibitor of metalloproteinase 4 (TIMP4), and Tenascin C (TNC)) potentially involved in the genetic basis of IA [37]. McColgan et al. conducted a meta-analysis that revealed associations between polymorphisms in the endothelial nitric oxide synthase (eNOS) and IL-6 genes and ruptured/unruptured IA, while the IL-6 G174C polymorphism showed a protective effect [38]. Researchers recently found that the gene serine protease inhibitor clade A member 3 (SERPINA3), which encodes a serine protease inhibitor, increases the risk of aneurysmal subarachnoid hemorrhage in Polish people [39]. However, Liu et al. did not find an association between the rs4934 polymorphism of SERPINA3 and sporadic IA in a Chinese population, highlighting potential ethnic variations [40]. GWASs have identified several SNPs associated with IA [41] (Table 1). Yasuno et al. demonstrated the association of SNPs within three intervals located at chromosomes 4q31.23, 12q22, and 20p12.1 with IA. The most significant locus, at 4q31.23, contains the endothelin receptor type A (EDNRA) gene, which may be involved in IA progression and rupture [42].
DNA methylation, an epigenetic modification involving the addition of a methyl group to DNA, can also influence disease susceptibility [43]. DNA methylation patterns are regulated by both genetic and environmental factors and play a crucial role in gene expression [27,44]. Disruptions in DNA methylation patterns have been linked to various diseases, including IA [27]. For example, Zhao et al. [45] suggested that the methylation level of mitogen-activated protein kinase 10 (MAP3K10) might serve as a predictive marker for IA risk, particularly in women. Similarly, Xu et al. [46] found glutathione S-transferase alpha 4 (GSTA4) mRNA expression and gender-specific DNA methylation in relation to IA. Zhou et al. [47] observed significantly higher levels of patatin-like phospholipase domain-containing protein 6 (PNPLA6) methylation in IA patients compared with the control group. Maimaiti et al.’s bioinformatics study [48] linked DNA methyltransferase 3 alpha (DNMT3A) to lower SAH and unruptured intracranial aneurysm (UIA) risks, whereas methyl-CpG-binding domain protein 2 (MBD2) was linked to higher UIA risks, highlighting the role that DNA methylation plays in the pathogenesis of IA.
Table 1. Potential IA nucleic acid biomarkers.
Table 1. Potential IA nucleic acid biomarkers.
ReferenceBiomarkerSample SourcenTime of TakeMethodologyIA vs.
Control
RIA vs. UIA
Zhong et al. [49]miR-205human blood91pretreatmentqRT-PCR
Feng et al. [50]miR-155-5pratsNApre/posttreatmentqRT-PCR
Yu et al. [51]miR-31a-5pratsNApre/posttreatmentqRT-PCR
Xiong et al. [52]miR-125ahuman blood50pretreatmentqRT-PCR
Holcomb et al. [53]miR-1rabbit6posttreatmentSequencing
miR-9-5prabbit6posttreatmentSequencing
miR-204-5prabbit6posttreatmentSequencing
miR-10a-5prabbit6posttreatmentSequencing
miR-21-5prabbit6posttreatmentSequencing
miR-34a-5prabbit6posttreatmentSequencing
miR-146a-5prabbit6posttreatmentSequencing
miR-223-3prabbit6posttreatmentSequencing
Jiang et al. [54]miR-1IA tissues14intraoperativeqRT-PCR
miR-7-1-3pIA tissues14intraoperativeqRT-PCR
miR-23b-5pIA tissues14intraoperativeqRT-PCR
miR-23b-3pIA tissues14intraoperativeqRT-PCR
miR-24-1-5pIA tissues14intraoperativeqRT-PCR
miR-28-5pIA tissues14intraoperativeqRT-PCR
miR-28-3pIA tissues14intraoperativeqRT-PCR
miR-29b-2-5pIA tissues14intraoperativeqRT-PCR
miR-29c-5pIA tissues14intraoperativeqRT-PCR
miR-29c-3pIA tissues14intraoperativeqRT-PCR
miR-133aIA tissues14intraoperativeqRT-PCR
miR-133bIA tissues14intraoperativeqRT-PCR
miR-140-3pIA tissues14intraoperativeqRT-PCR
miR-143-5pIA tissues14intraoperativeqRT-PCR
miR-143-3pIA tissues14intraoperativeqRT-PCR
miR-145-5pIA tissues14intraoperativeqRT-PCR
miR-145-3pIA tissues14intraoperativeqRT-PCR
miR-455-5pIA tissues14intraoperativeqRT-PCR
Zhao et al. [55]miR-29ahuman blood24pre/posttreatmentqRT-PCR
Zheng et al. [56]miR-513b-5phuman serum100pretreatmentqRT-PCR
Fan et al. [57]miR-331-3pIA tissues96intraoperativeRT-qPCR
Wang et al. [25]miR-29ahuman plasma165pretreatmentqRT-PCR
Bekelis et al. [58]miR-21IA tissues7intraoperativesequencing
miR-143-5pIA tissues7intraoperativesequencing
miR-145IA tissues7intraoperativesequencing
Lv et al. [59]miR-136-5pIA tissues82intraoperativeqRT-PCR
Cai et al. [60]miR-92aIA tissues90intraoperativeqRT-PCR
Liu et al. [61]miR-29bIA tissues6intraoperativeqRT-PCR
Guo et al. [62]miR-23b-3pIA tissues32intraoperativeqRT-PCR
Jin et al. [63]miR-22human plasma24pretreatmentPCA
miR-671-5phuman plasma24pretreatmentPCA
miR-720human plasma24pretreatmentPCA
miR-365human plasma24pretreatmentPCA
miR-498human plasma24pretreatmentPCA
miR-574human plasma24pretreatmentPCA
miR-106bhuman plasma24pretreatmentPCA
miR-21human plasma24pretreatmentPCA
miR-936human plasma24pretreatmentPCA
Su et al. [64]miR-132human plasma58pretreatmentqRT-PCR
miR-324human plasma58pretreatmentqRT-PCR
Supriya et al. [65]miR-26bIA tissues29intraoperativeqRT-PCR
miR-199aIA tissues29intraoperativeqRT-PCR
miR-497IA tissues29intraoperativeqRT-PCR
miR-365IA tissues29intraoperativeqRT-PCR
Luo et al. [66]miR-9IA tissues13intraoperativeqRT-PCR
Yang et al. [67]miR-144-5phuman plasma84pretreatmentqRT-PCR
Liao et al. [68]miR-145-5phuman plasma12pretreatmentqRT-PCR
miR-29a-3phuman plasma12pretreatmentqRT-PCR
Li et al. [69]miR-16human plasma40pretreatmentqRT-PCR
miR-25human plasma40pretreatmentqRT-PCR
Zou et al. [70]miR-34ahuman blood20intraoperativeqRT-PCR
Xu et al. [71]miR-143human serum30intraoperativeqRT-PCR
miR-145human serum30intraoperativeqRT-PCR
Yuan et al. [72]miR-34ahuman serum20intraoperativeqRT-PCR
Zhao et al. [45]Methyl MAP3K10human plasma96pretreatmentMIRA-seq
Xu et al. [46]Methyl GSTA4human blood44pretreatmentMIRA-seq
Zhou et al. [47]Methyl PNPLA6human plasma96pretreatmentMIRA-seq
Roder et al. [37]BCL2/30pretreatmentMIRA-seq
COL1A2/30pretreatmentMIRA-seq
COL3A1/30pretreatmentMIRA-seq
COL5A2/30pretreatmentMIRA-seq
CXCL12/30pretreatmentMIRA-seq
TIMP4/30pretreatmentMIRA-seq
TNC/30pretreatmentMIRA-seq
Maimaiti et al. [48]DNMT3A/156pretreatmentMIRA-seq
MBD2/156pretreatmentMIRA-seq
Abbreviations: IA, intracranial aneurysm; UIA, unruptured intracranial aneurysm; RIA, ruptured intracranial aneurysm; qRT-PCR, quantitative real-time polymerase chain reaction; PCA, principal component analyzed; MIRA-seq, methylated-pyrosequencing; Methyl, methylation; MAP3K10, mitogen-activated protein kinase 10; GSTA4, glutathione S-transferase alpha 4; PNPLA6, patatin-like phospholipase domain-containing protein 6; BCL2, B-cell lymphoma-2; COL1A2, collagen type I alpha 2 chain; COL3A1, collagen type III alpha 1 chain; COL5A2, collagen type V alpha 2 chain; CXCL12, chemokine (C-X-C motif) ligand 12; TIMP4, tissue inhibitor of metalloproteinase 4; TNC, Tenascin C; DNMT3A, DNA methyltransferase 3 alpha; MBD2, methyl-CpG-binding domain protein 2; ↑, upregulate; ↓, downregulate.

6. RNA

MicroRNAs (miRNAs) are small, non-coding RNAs, typically consisting of approximately 22 nucleotides, that regulate gene expression through transcription or translation interference [28]. They play crucial roles in various physiological processes, including cell cycle progression, stem cell differentiation, organ growth, and signal transduction [73]. miRNAs are also involved in tumorigenesis, regulation of oxidative stress, DNA damage, and the organismal response to radiation [74]. According to several studies, miRNAs play a crucial role in IA formation and rupture (Figure 3) (Table 1). Bekelis et al. [58] identified differentially expressed genes by analyzing tissue specimens from IA and control subjects. Matrix metalloproteinase-13 (MMP-13) and various collagen genes [ collagen type I alpha 1 chain (COL1A1), collagen type V alpha 1 chain (COL5A1) and alpha 2 chain (COL5A2) ], and miR-21 showed the greatest upregulation, while miR-143-5p showed the greatest downregulation. Jiang et al. [54] used microarrays to compare miRNA expression between IA tissue and normal tissue. They identified 18 significantly differentially expressed miRNAs, including miR-1, miR-24-1-5p, miR-29b-2-5p, miR-7-1-3p, miR-29c-5p, miR-29c-3p, miR-133a, miR-133b, miR-23b-3p, miR-140-3p, miR-143-5p, miR-143-3p, miR-23b-3p, miR-145-5p, miR-145-3p, miR-455-5p, and others. Functional analysis revealed that the predicted target genes of these miRNAs were associated with processes such as macrophage migration, monocyte proliferation, and smooth muscle cell migration, which are important in IA development. Fan et al. [57] analyzed the IA microarray GSE75436 from the gene expression omnibus (GEO) database, which included normal and IA samples. Their analysis revealed significantly decreased miR-331-3p expression in IA. They proposed that miR-331-3p plays a role in IA by regulating the VSMC phenotype and inhibiting TNF-α and CD14 through the NF-κB signaling pathway, indicating its potential as a therapeutic target and prognostic marker. Compared with microarrays, next-generation RNA sequencing (RNA-seq) offers higher specificity, sensitivity, and a wider dynamic range. It can also detect novel transcripts and isoforms [75]. Zhong et al. found that miRNA-205 levels in the blood of IA patients were significantly higher than those in the healthy control group, suggesting its potential as a diagnostic biomarker [49]. Conversely, Xiong et al. [52] observed elevated levels of miR-125a in the blood of IA patients compared with the control group, potentially increasing the risk of IA rebleeding. Interestingly, miR-125a is usually downregulated in certain cancers, suggesting complex regulatory mechanisms. Holcomb et al. [53] identified miRNAs that were upregulated (miR-10a-5p, miR-21-5p, miR-34a-5p, miR-146a-5p, and miR-146a-5p) and downregulated (miR-1, miR-9-5p, and miR-204-5p) in arterial aneurysm tissues. Pathway analysis suggested that some upregulated miRNAs might promote inflammation, while the downregulated miRNAs might inhibit cell migration and coagulation, affecting aneurysm development. Zhao et al. [55] found that miR-29a is significantly upregulated in IA tissues and promotes apoptosis, leading to increased DNA damage and mitochondrial release. It may also reduce the expression of myeloid leukemia 1 (Mcl-1) and increase the expression of caspase-3 and cytochrome c, promoting the activation of the apoptosis pathway. These findings suggest that miR-29a could be a novel target for IA prevention and treatment. Consistent with these results, Wang et al. [25] also found miR-29a to be a potential biomarker for IA. Zheng et al. [56] found that there was a decrease in miR-513b-5p expression in both unruptured and ruptured IA patients compared with the healthy control group. Their experiments suggest that miR-513b-5p targets COL1A2 and COL1A1 to inhibit smooth muscle cell proliferation and promotes cell death and apoptosis. This aligns with previous studies suggesting an anti-proliferative and pro-apoptotic role for miR-513b-5p [76]. Yuan et al. [72] observed that serum levels of miR-34a were significantly decreased in IA patients compared with the control group, suggesting a potential role for miR-34a in IA formation and progression and potentially serving as a biomarker for early detection. Yang et al. found that serum levels of extracellular vesicle miR-144-5p were significantly lower in IA patients compared with healthy individuals [67]. Conversely, Liao et al. reported elevated levels of miR-145-5p and miR-29a-3p in circulating EVs from IA patients compared with controls, suggesting their potential use for monitoring IA formation and rupture [68]. However, Xu et al. [71] observed downregulation of miR-143/145 in the serum of IA patients, contradicting Liao et al.’s findings [68]. This discrepancy highlights the complexity of miRNA regulation in IA and the need for further investigation. MiR-145 is known to regulate VSMC phenotype and proliferation, potentially impacting IA pathogenesis [71]. In vitro experiments suggest that miR-143/145 may counteract the regulatory effects of Kruppel-like factor 5 (KLF5) in VSMCs, influencing cell proliferation and contraction in IA. The differing results observed by Liao et al. [68] and Xu et al. [71] could be due to compensatory mechanisms at play [71]. Li et al. identified miR-16 and miR-25 as potential circulating miRNA biomarkers for IA using microarray profiling and validation with quantitative real-time polymerase chain reaction (qRT-PCR) [69]. Feng et al. proposed that tumor-associated macrophage (TAM)-derived exosomal miR-155-5p promotes IA formation by antagonizing gremlin 1 (GREM1), a secreted bone morphogenetic protein antagonist. This suggests that miR-155-5p may serve as a therapeutic target and a biomarker for IA [50].
RNA modifications, including N1-methyladenosine (m1A), N6-methyladenosine (m6A), m7G, 2′-O-methylation, 5-methylcytosine (m5C), and ac4C RNA acetylation, represent another layer of epigenetic regulation influencing RNA metabolism and processing [77]. m6A is considered the most abundant form. m6A modification occurs on adenosine within the RRACH sequence and is regulated by “Writer”, “Eraser”, and “Reader” complexes [78]. Mounting evidence suggests the crucial role of m6A methylation in various diseases, including cancer [79]. However, limited research has been conducted on m6A methylation in IA. Maimaiti et al. [80] analyzed 60 samples (44 IA, 16 normal) and identified 8 m6A markers (IGFBP2, IGFBP1, IGF2BP2, YTHDF3, ALKBH5, RBM15B, LRPPRC, and ELAVL1) that correlated significantly with IA, suggesting their potential as prognostic biomarkers. Li et al. [81] analyzed three datasets and found nine differentially expressed m6A regulators (insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), zinc finger protein 217 (ZNF217), YT521-B homology domain family member 2 (YTHDF2), YT521-B homology domain family member 3 (YTHDF3), YT521-B homology domain-containing protein 1 (YTHDC1), fat mass and obesity-associated protein (FTO), RNA-binding motif protein 15 (RBM15), and leucine-rich pentatricopeptide repeat containing (LRPPRC)) between the control and IA groups, also indicating their potential as biomarkers. However, these studies have limitations. The small sample sizes may lead to bias, and the validation of m6A regulatory factor expression was limited to blood samples and did not encompass IA tissues. Additionally, specific m6A sites have not been investigated using techniques like MeRIP-seq and MeRIP-qPCR. These techniques could help identify the target genes of m6A regulatory factors that play a role in IA development.

7. Proteins

7.1. Inflammation-Related Proteins

Inflammation, a natural response to injury characterized by swelling, redness, and increased white blood cell infiltration, plays a dual role in the context of IA. While it promotes initial tissue repair, chronic inflammation can lead to detrimental effects [82]. The development of IA involves distinct stages: endothelial injury and elastic layer degradation, recruitment and infiltration of inflammatory cells, and chronic vascular wall remodeling [29]. Inflammatory cell infiltration is a crucial step, as these cells secrete inflammatory cytokines that regulate, activate, and promote the proliferation, migration, and apoptosis of immune and endothelial cells. Additionally, they release matrix-degrading collagenases and proteases, which contribute to aneurysm formation and progression [29]. This section highlights the role of key inflammatory-related proteins in IA (Table 2).
Monocyte chemoattractant protein-1 (MCP-1), a chemokine expressed by microglia, neurons, astrocytes, and endothelial cells, serves as a critical player in monocyte recruitment [83]. It promotes monocyte aggregation in damaged areas by regulating adhesion molecules and modulating membrane-binding activity. MCP-1 also induces the release of molecules that degrade elastic and collagen fibers in the arterial wall (e.g., MMPs), leading to aneurysm formation [83]. Under high shear stress conditions within the aneurysmal wall, endothelial cells express MCP-1, facilitating early-stage macrophage infiltration [12]. Studies have shown that MCP-1 levels are elevated in ruptured IAs compared with unruptured IAs, while miR-493, a competitive endogenous RNA of AC007362 and a direct target gene of miR-493, exhibits an inverse relationship with MCP-1 expression [31,84]. These findings suggest that MCP-1 may serve as a biomarker for IA rupture. Additionally, in IA patients, MCP-1 levels were observed to be elevated in comparison with those in healthy controls, with further increases observed in patients with ruptured or multiple aneurysms [82,85]. These results highlight the critical role of MCP-1 in IA formation, development, and rupture. Liu et al. [86] observed a significant increase in MCP-1 expression in an IA model, suggesting continuous production by SMCs and endothelial cells, potentially contributing to a sustained inflammatory response. Notably, MCP-1 expression may serve as an early indicator for IA detection, as its mRNA levels rapidly peak within a week after induction [86].Tumor necrosis factor-alpha (TNF-α) is a pro-inflammatory cytokine involved in the inflammatory response cascade, and it plays a role in initiating cell apoptosis signaling pathways [87]. Produced by macrophages, endothelial cells, and SMCs within IA, TNF-α contributes to pro-inflammatory changes [88]. It acts as a pro-inflammatory factor in IAs, inducing macrophage polarization and promoting atherosclerosis development [89]. TNF-α exerts its pro-atherosclerotic effects through various mechanisms, including disrupting endothelial junctions (leading to dysfunction and instability) and facilitating leukocyte migration by stimulating adhesion molecules, particularly Vascular Cell Adhesion Molecule 1 (VCAM-1) [87,88]. Kanematsu et al. [90] demonstrated that TNF-α directly activates genes in cerebral vascular SMCs associated with IA formation, including matrix metalloproteinase-3(MMP-3), matrix metalloproteinase-9(MMP-9), MCP-1, VCAM-1, and IL-1β, all involved in pro-inflammatory responses and matrix remodeling. Fan et al. [57] revealed increased expression of TNF-α, ICAM-1, MCP-1, IL-6, matrix metalloproteinase-2 (MMP-2), and MMP-9 in IA patient vessels compared with controls. These findings support the role of TNF-α in promoting inflammation and matrix remodeling in IA. Similar to MCP-1, TNF-α levels are elevated in both unruptured and ruptured IAs compared with controls, with higher levels observed in ruptured IAs, suggesting its potential role in promoting both formation and rupture [85]. Starke et al. reported increased TNF-α expression in a mouse model of IA, with further elevation in ruptured aneurysms [91]. Additionally, TNF-α gene knockout mice and mice pretreated with TNF-α inhibitors displayed a decreased incidence of IA formation and rupture, suggesting its involvement in these processes [91]. However, the precise role of TNF-α in aneurysm progression and rupture remains to be elucidated. Jayaraman et al. observed increased expression of TNF-α and its downstream apoptotic target fatty acid synthase (FAS) in IA, suggesting its role in promoting apoptosis and inflammation within the aneurysm wall [92]. TNF-α may also serve as a potential biomarker for IA formation and rupture.
Table 2. Potential IA protein biomarkers.
Table 2. Potential IA protein biomarkers.
ReferenceBiomarkerSample SourcenTime of TakeMethodologyIA vs.
Control
RIA vs. UIA
Xiong et al. [52]ET-1blood50pretreatmentWB
Lv et al. [59]KDM1AIA tissues82intraoperativeWB
Cai et al. [60]HDAC9IA tissues90intraoperativeWB
BCL2L11IA tissues90intraoperativeWB
Fan et al. [57]TNF-αIA tissues96intraoperativeWB
CD14IA tissues96intraoperativeWB
Zheng et al. [56]COL1A1serum100pretreatmentWB
COL1A2serum100pretreatmentWB
TNF-αserum100pretreatmentWB
IL-1βserum100pretreatmentWB
MMP-2serum100pretreatmentWB
MMP-3serum100pretreatmentWB
MMP-9serum100pretreatmentWB
Guo et al. [62]PTENIA tissues32intraoperativeWB
Xu et al. [46]GSTA4serum44pretreatmentWB
Zhou et al. [47]PNPLA6serum96pretreatmentWB
Zhang et al. [82]IL-1βblood66pretreatmentBio-Plex protein array systems
MCP-1blood66pretreatmentBio-Plex protein array systems
TNF-αblood66pretreatmentBio-Plex protein array systems
Yamaguchi et al. [93]MMP-3IA tissues24pre/intraoperativeWB
IL-1βIA tissues24pre/intraoperativeWB
Zou et al. [70]MMP-2IA tissues20pre/intraoperativeWB
Kamińska et al. [94]MCP-1CSF25pretreatmentELISA
IL-8CSF25pretreatmentELISA
Lai et al. [85]TNF-αserum108pretreatmentELISA
MCP-1serum108pretreatmentELISA
IL-1βserum108pretreatmentELISA
IL-6serum108pretreatmentELISA
NF-κB p65 serum108pretreatmentELISA
Kamińska et al. [95]NF-κB p65 CSF, serum25pretreatmentELISA
CXCL1CSF, serum25pretreatmentELISA
CXCR2CSF, serum25pretreatmentELISA
Aoki et al. [96]COX-2mice IA tissues5pretreatmentWB
mPGES1mice IA tissues5pretreatmentWB
Sun et al. [97]NOX4IA tissues27pretreatmentWB
p22phoxIA tissues27pretreatmentWB
p47phoxIA tissues27pretreatmentWB
TRPC6IA tissues27pretreatmentWB
CNIA tissues27pretreatmentWB
NFATC1IA tissues27pretreatmentWB
MMP-2IA tissues27pretreatmentWB
MCP-1IA tissues27pretreatmentWB
Aoki et al. [98]procollagen type IIA tissues6pretreatmentWB
procollagen type IIIIA tissues6pretreatmentWB
Abbreviations: ET-1, endothelin-1; KDM1A, lysine-specific demethylase 1; HDAC9, Histone deacetylase 9; BCL2L11, BCL2 (B-cell lymphoma-2)-like protein 11; PTEN, phosphatase and tensin homolog; GSTA4, glutathione S-transferase alpha 4; PNPLA6, patatin-like phospholipase domain-containing protein 6; NF-κB p65, Nuclear factor kappa-B p65 subunit; CXCL1, C-X-C motif ligand 1; CXCR2,C-X-C Motif Chemokine Receptor 2; TRPC6,The transient receptor potential canonical 6; CN, Calcineurin; NFATC1, nuclear factor of activated T cell; TNF-α, tumor necrosis factor-α; CD14, cluster of differentiation 14; COL1A1, collagen type I alpha 1 chain; COL1A2, collagen type I alpha 2 chain; IL-1β, interleukin-1β; MMP-2, Matrix Metallopeptidase 2; MMP-3, Matrix Metallopeptidase 3; MMP-9, Matrix Metallopeptidase 9; MCP-1, monocyte chemotactic protein-1; IL-8, interleukin-8; IL-6, interleukin-6; COX-2, Cyclooxygenase-2; mPGES1, microsomal prostaglandin E synthase-1; NOX4, NADPH oxidase 4; p22phox, Cytochrome b-245, alpha polypeptide; IA, intracranial aneurysm; UIA, unruptured intracranial aneurysm; RIA, ruptured intracranial aneurysm; CSF, cerebrospinal fluid; WB, Western blotting; ELISA, Enzyme-linked immunosorbent assay; p47phox, Cyclin-dependent kinase inhibitor p27; ↑, upregulate; ↓, downregulate.
IL-1β is a pro-inflammatory cytokine produced in its precursor form by activated macrophages and processed into its active form by caspase 1 (CASP1/ICE). It is produced by various cell types, including monocytes, macrophages, T cells, B cells, dendritic cells, endothelial cells, and VSMCs [99]. IL-1β plays a crucial role in inflammatory responses, influencing cell proliferation, differentiation, and apoptosis. Studies have shown that IL-1β contributes to inflammatory pain hypersensitivity by inducing cyclooxygenase-2 (PTGS2/COX2) in the central nervous system (CNS) [100]. Animal models of IAs and abdominal aortic aneurysms (AAAs) suggest that IL-1β upregulation promotes aneurysm progression by regulating collagen biosynthesis in the aneurysm wall [101]. In a study by Moriwaki, T. et al., IL-1β−/− mice displayed a significantly lower number of late-stage aneurysms compared with wild-type mice, with a notable reduction in cellular apoptosis in the aneurysm tissue [102]. These findings suggest a protective role for IL-1β deficiency against late-stage aneurysm development. Zheng et al. reported there was a notable rise in IL-1β mRNA expression among IA patients., with further elevation observed in ruptured compared with unruptured IAs [56]. They proposed that necrotic VSMCs in aneurysms can release IL-1β, inducing IL-6 production, thereby promoting vascular inflammation and accelerating aneurysm progression. Additionally, Zhang et al. [82] and Lai et al. [85] found elevated IL-1β levels in IA patients, suggesting its importance in aneurysm development and rupture. Liu et al. [103] investigated the correlation between arterial wall enhancement and cellular apoptosis. They found that patients with unruptured IAs showing arterial wall enhancement had upregulated pro-inflammatory cytokines like IL-1β and downregulated anti-inflammatory cytokines like IL-1ra. This suggests that the ratio of IL-1β to IL-1ra might serve as a potential biomarker for predicting aneurysm rupture or growth.
NF-κB is a transcription factor that regulates various inflammation-related genes in response to external stimuli. The NF-κB protein family consists of two main classes: class I (including NF-κB1/p105 and NF-κB2/p100) and class II (including RelA/p65, RelB, and c-Rel) [104]. NF-κB signaling involves two interconnected pathways, the canonical pathway and the non-canonical pathway, which differ in their components and functions [104]. In mammals, NF-κB forms homodimers or heterodimers from five subunits: p50, p52, RelA (p65), RelB, and c-Rel. Aoki et al. [105] demonstrated the crucial role of the p50 subunit in initiating IA formation. They found that p50−/− mice exhibited reduced expression of MCP-1 and VCAM-1, as well as decreased macrophage infiltration in the cerebral artery wall, compared with p50+/+ mice after aneurysm induction. This suggests that p50 plays a key role in macrophage aggregation and potentially in degenerative changes in the aneurysm wall through transcriptional regulation. Smoking, alcohol abuse, obesity, and oxidative stress are potential risk factors for IA formation and rupture. These factors can activate NF-κB, leading to increased expression of pro-inflammatory markers and MMPs [106]. In its inactive state, NF-κB resides in the cytoplasm. Upon stimulation, it becomes activated, translocates to the nucleus via nuclear localization sequences, and initiates the regulation of target genes [107]. Liu et al. [86] observed strong nuclear expression of chemokines, adhesion molecules, MMPs, and other factors in aneurysmal cells, indicating NF-κB activation. Activated NF-κB promotes the expression of genes encoding chemokines, adhesion molecules, MMPs, etc., which directly influence the activation, proliferation, migration, and secretion processes of relevant cells [108]. These findings suggest that NF-κB plays a critical role in aneurysmal wall inflammation and the vascular pathology of IA. Interestingly, Lai et al. [85] detected increased NF-κB p65 protein expression in both unruptured and ruptured IA compared with normal vessels, suggesting its potential role as a risk factor. However, Kamińska et al. [95] reported lower NF-κB p65 concentrations in the serum and cerebrospinal fluid (CSF) of unruptured IA patients compared with controls, suggesting potential increased activation within the central nervous system. These findings suggest that NF-κB plays a complex role in IA, potentially influencing both initiation and progression. Further research is needed to elucidate the precise contribution of different NF-κB subunits and pathways at various stages of IA development.
IL-6, a multifunctional cytokine crucial for host defense, exerts distinct effects in the nervous system through classical and trans-signaling pathways [109]. Both pathways involve binding to either membrane-bound or soluble IL-6 receptors, which activate JAK/STAT and MAPK pathways, ultimately leading to gene transcription and protein expression [110,111]. The classical pathway, primarily mediated by membrane-bound receptors, has neuroprotective effects. Upon binding to glycoprotein 130 (gp130), IL-6 activates the JAK/STAT and MAPK pathways, promoting cell growth, survival, and gene expression [110,111]. This pathway additionally results in the generation of cytokine signaling suppressor 3 (SOCS3), an IL-6 signaling negative regulator, creating a feedback loop to limit further IL-6 activity [112]. Conversely, the trans-signaling pathway, involving soluble receptors, promotes inflammation. Soluble IL-6 receptors bind to gp130 on endothelial cells, activating similar JAK/STAT and MAPK pathways but in a wider range of cell types. This pathway also inhibits SOCS3, leading to sustained IL-6 signaling and increased production of inflammatory cytokines [112,113]. The role of IL-6 in IA development and progression remains to be fully elucidated. Animal studies suggest a potential link between IL-6 and IA formation. Reduced cytokine production, including IL-6, in macrophages following dipeptidyl peptidase-4 (DPP-4) inhibition correlated with smaller IA size in a rat model [114]. Clinical studies have explored IL-6 levels in IA patients. Wajima et al. [115] observed higher IL-6 expression in IA tissues compared with controls, with potentially higher levels in women. Kamińska et al. [116] reported an elevated IL-6 ratio (CSF to serum concentration) in unruptured IA patients, suggesting potential central nervous system involvement, especially in those with multiple aneurysms. Additionally, Kao et al. [26] identified elevated blood IL-6 within the first three days of IA rupture as an independent predictor of poor clinical outcomes. Interestingly, animal models of SAH, a complication of ruptured IA, suggest that IL-6 may contribute to vasospasm, a narrowing of blood vessels that can worsen neurological deficits [117]. These discoveries imply that IL-6 might have a complex function in various aspects of IA, potentially influencing the formation, growth, rupture, and post-rupture complications. Further research is needed to determine the precise role of IL-6 and its signaling pathways in IA pathogenesis.

7.2. Oxidative Stress-Related Proteins

Oxidative stress, resulting from an imbalance between the generation and removal of free radicals, plays a pivotal role in the pathogenesis of IA. This imbalance leads to DNA damage, cellular toxicity, and apoptosis [118]. In the pathogenesis of IA, crucial pathways of oxidative stress involve atherosclerosis, hemodynamic stress, endothelial dysfunction, VSMC phenotype modulation, vascular remodeling, and apoptotic cell death. These pathways promote the generation of reactive oxygen species (ROS) such as superoxide (O2•−), hydrogen peroxide (H2O2), and peroxynitrite (ONOO•−) [119]. Recognized risk factors contributing to ROS production include cigarette smoke, alcohol, and hypertension (Figure 4). Smoking is a primary risk factor for IA formation and rupture, with approximately 80% of SAH patients reporting a significant smoking history [120]. Cigarette smoke is a major source of free radicals, activating Nicotinamide Adenine Dinucleotide Phosphate (NADPH) and generating O2•− and H2O2. These may originate directly from gas/tar phases, activated white blood cells, or endogenous sources of ROS, such as xanthine oxidase, uncoupled eNOS, and the mitochondrial electron transport chain [121]. Alcohol is widely recognized as another significant risk factor for aneurysm rupture, with its effects potentially mediated through free radicals. However, further extensive research is needed to determine whether alcohol-induced oxidative stress directly leads to aneurysmal changes in cerebral vasculature [122]. Hypertension has been demonstrated as another risk factor for the formation and rupture of aneurysms [122]. Evidence suggests that O2•−, H2O2, and ONOO•− generated through NADPH oxidase and uncoupled eNOS may play a crucial role in hypertension pathogenesis, although various other factors may also be involved [123]. The potential sources of oxidative stress in IA formation and rupture are numerous. The main enzymatic sources of ROS in the cerebrovascular system include COX, lipoxygenases (LOXs), and NADPH oxidase [124]. NOS is another potential source of free radicals in the cerebral circulation [125]. Oxidative stress-related proteins play an indispensable role in the process of IA formation and rupture (Table 2).
Cyclooxygenase-2 (COX-2), an inducible enzyme unlike the stable COX-1, plays a significant role in IA development. Studies have shown increased COX-2 expression in IA tissues compared with healthy controls [96]. This upregulation is likely due to the presence of unstable sequences in the COX-2 gene (Ptgs-2), making it readily activated during inflammatory processes [126,127]. A key function of COX-2 is the conversion of arachidonic acid to prostaglandin G2 (PGG2), a precursor for various inflammatory mediators. In the context of IA, the prostaglandin E2 (PGE2) derived from COX-2 activity appears to be particularly crucial [128]. PGE2 signaling through the EP2 receptor activates NF-κB, a transcription factor promoting further COX-2 expression. This positive feedback loop involving COX-2, PGE2, and NF-κB is believed to be a key driver of inflammation in the IA wall [96]. Therefore, targeting the COX-2 pathway may hold promise for therapeutic strategies aimed at preventing or mitigating IA formation.
Typical NADPH oxidase (NOX) is primarily expressed in polymorphonuclear neutrophils (PMNs), playing a central role in host defense and participating in bacterial killing by PMN. The enzyme consists of two membrane-bound subunits (gp91phox and p22phox) and at least three cytosolic subunits [129]. NOX is widely distributed in the vasculature, including endothelial cells, VSMCs, and adventitial cells of cerebral arteries, exhibiting active functionality. Various factors, including cytokines and mechanical stress, can induce vascular NOX to produce superoxide and hydrogen peroxide, contributing to oxidative stress in IA [130]. Tamura et al. [131] demonstrated in rat experiments that estrogen may mediate the expression of NADPH oxidase in aneurysms, providing a potential link between oxidative stress and the increased incidence of aneurysms in women. In VSMCs, the isoform NOX4 of NOX is the predominant form [132]. Studies have shown that ROS derived from NOX4 can induce phenotypic transition of human brain vascular smooth muscle cells (HBVSMCs) and promote the formation of IAs [133]. Sun et al. [97] found that the expression of NOX4 increased in IA patients, and the TRPC6-NFATC1 signaling was activated in IA patients. Their study demonstrated that interruption of TRPC6-NFATC1 signaling significantly downregulated NOX4 expression in IA and attenuated VSMC phenotypic transition, suggesting that TRPC6-NFATC1 signaling could be an important therapeutic target for treating IA. Intracellular Ca2+ imbalance mediated by transient receptor potential-6 (TRPC6) may also participate in the regulation of NOX4 and the VSMC phenotypic switch in the pathogenesis of IA.
The nitric oxide synthase (NOS) enzyme family consists of eNOS, located in endothelial cells; inducible NOS (iNOS), located in macrophages and VSMCs; and neuronal NOS (nNOS), located in neurons. NOS primarily synthesizes nitric oxide (NO), a hydrophobic molecule with a short half-life, which easily diffuses through neighboring cell membranes, exerting transient effects locally [134]. In vascular physiology, nitric oxide plays various crucial roles, including regulating vascular tone, inhibiting SMC proliferation, suppressing pro-inflammatory mediators, and maintaining the proper function and integrity of endothelial cells (ECs) [135]. Studies have found that while eNOS and nNOS have protective effects on IA, under abnormal conditions such as mechanical injury and inflammation, macrophages and VSMCs express iNOS [136]. ROS and inflammatory mediators (such as Interferon-gamma (IFN-γ), TNF-a, and IL-1b) increase the expression of iNOS, which is associated with the cytotoxic effects of NO. iNOS also impairs the activity of eNOS and nNOS through a negative feedback loop involving NO [119]. eNOS is primarily regulated by Ca2+–calmodulin binding, although it can also respond to WSS activation through phosphorylation by protein kinase B [137]. Acute or prolonged high laminar shear stress leads to the increased transcription and activation of eNOS, thereby promoting vasodilation to prevent initial vascular injury. However, under pathologically low WSS conditions, ECs fail to express eNOS [138]. Khurana et al. demonstrated, using gene chip technology, that the intron-4 27-base-pair variable number tandem repeat polymorphism (eNOS 27 VNTR) in the eNOS gene can predict the susceptibility to rupture of IAs [139]. In another study, Khurana et al. used gene microarray technology for detection, and the data results indicate that the eNOS T-786C genotype may be a factor influencing the size of aneurysm rupture in populations with known IA [140]. Therefore, NOS may be an important source of reactive nitrogen species (RNS) and ROS in IA, potentially promoting aneurysm formation and progression through various mechanisms [141].

7.3. Extracellular Matrix-Associated Proteins

The extracellular matrix (ECM) is a complex three-dimensional network composed of fibers, gels, and minerals, constituting the fundamental supportive structure for cell survival within biological tissues [142]. Under normal physiological circumstances, cells constantly modify the ECM through processes such as rearrangement, degradation, chemical modification, and synthesis [143]. However, in pathological conditions, the ECM undergoes notable remodeling in reaction to various stimuli, involving the removal of structural and functional proteins, playing a critical role in the formation of IA (Table 2).
Collagen is the main component of the ECM, playing a crucial role in offering structural support and maintaining tissue morphology and mechanical characteristics. It regulates cell growth, activity, and specialized development [144]. Collagen degradation is primarily regulated by MMPs. MMPs are accountable for the degradation of collagen types I, II, and III. Proteases including MMP-1 (interstitial collagenase), MMP-13 (collagenase-3), MMP-8 (neutrophil collagenase), and membrane-bound MMP-14 have the capability to degrade these collagen types. Specifically, MMP-2 is primarily responsible for degrading type I collagen; in contrast, MMP-13 is the enzyme of choice for degrading type II collagen, and MMP-8 and MMP-1 are mainly involved in the degradation of types I and III collagen [145]. The decrease in collagen content in the arterial wall is one of the prominent pathological features of IA [146]. Aoki et al. found that there was a decrease in collagen protein biosynthesis in the arterial aneurysm wall, representing an important aspect of the degenerative changes in the aneurysm wall. At the transcriptional level, the expression of type I and III procollagen in the aneurysm wall was downregulated [98]. In another study, they demonstrated that excessive collagen degradation in the wall of intracranial aneurysms is caused by an imbalance between MMPs and TIMPs as well as tissue proteases and cysteine protease C [147].
Elastin originates from tropoelastin, a soluble monomer crucial for maintaining the morphology, elasticity, and functionality of the ECM. It plays a critical supportive role physiologically [148]. Watton et al.’s aneurysm model suggests that aneurysm formation stems from the local degradation of elastin, leading to disturbances in arterial geometry. Further degradation of elastin is explicitly associated with low wall shear stress in specific regions of the artery, thereby promoting aneurysm development [149]. Nakagawa et al.’s analysis suggests that compared with unruptured aneurysms, the plasma concentration of soluble elastin fragments in the lumen of ruptured aneurysms is significantly higher [150]. A large-scale retrospective study in China reported that two single-nucleotide polymorphisms in the elastin gene are associated with the development and rupture of IAs, further emphasizing the role of elastin in aneurysm rupture [151]. Onda et al. found a strong association between variants in the elastin gene’s 20th intron/23rd intron (located on chromosome 7q11) and IA in Japan [152]. The measurement of elastin concentration in peripheral blood may be used to predict the risk of rupture in unruptured intracranial aneurysms.
Matrix metalloproteinases (MMPs) are a class of proteinases involved in the regulation of ECM by degrading proteins, regulating cell adhesion, and modulating cytokines [153]. MMPs were first discovered in 1962 and were initially identified as collagenases mediating the absorption of tadpole tails. This protein family shares structural similarities, and its enzymatic activity facilitates a range of biological processes. [154]. Most proteins found in the ECM are substrates of MMPs. MMPs and their degradation products have the ability to alter the downstream effects of cellular signaling molecules such as growth factors, cell adhesion molecules, cytokines, and other MMPs [155]. The regulation of MMP expression involves multiple factors, including inflammatory cytokines like TNF-α and IL-1β, vascular factors such as endothelin A, and ECM proteins [156]. TIMPs are the primary endogenous inhibitors of MMP activity, exerting their effects through binding to the C-terminal domain of MMPs [157]. In the development and pathogenesis of IA, MMPs and TIMPs play critical roles. MMPs belong to the protease family involved in vascular remodeling. Elevated activity of MMP-2, MMP-3, and MMP-9 within the arterial wall contributes to inflammation and remodeling of the extracellular matrix during IA [158]. Kim et al. used GWAS data to investigate the association between MMPs and IA. Three SNPs of MMP-24 were discovered to be significantly linked to IA, exerting a protective influence on IA development. They speculated that MMP-24 could impede IA formation by influencing ECM stiffness. Regarding MMP-13, they found a close association with IA, but it is unclear whether it facilitates or impedes IA formation. Additionally, they discovered MMP-2 to be the MMP most strongly associated with IA, with high expressions in vasculature, brain, and blood [159]. Aoki et al. reported that the elevated levels of MMP-2 and MMP-9, derived from macrophages within the IA wall, are closely associated with the progression of aneurysms [160]. Liu et al. found that in experimental cerebral aneurysms, the elastic fibers of the extracellular matrix were disrupted, leading to a reduction or loss of elastic fibers accompanied by extensive infiltration of inflammatory cells. The expression of MMP-9 in the aneurysm wall was significantly enhanced, and positive expression of MMP-9 was observed throughout the aneurysm wall. These results suggest that damage to elastic fibers is one of the key factors in aneurysm formation, while increased infiltration of inflammatory cells and secretion of MMP-9 are the main causes of elastic fiber damage [86]. The mechanism responsible for the increase in MMP levels during the development and rupture of IA remains uncertain. Zheng et al. found that compared with the control group, the mRNA expression levels of TNF-α, MMP-2, MMP-3, and MMP-9 were notably higher in IA patients. By delving deeper into the regulatory function of miR-513b-5p in the RIP1/RIP3/MLKL and MMP pathways in IA, they found that miR-513b-5p enhanced TNF-α-induced expression of MMP-2 and MMP-9 proteins. Additionally, they also discovered that the expression of TNF-α and MMP-9 was markedly elevated in patients with ruptured IAs in comparison with those with unruptured IAs. They concluded that TNF-α and MMP-9 have a greater impact on the rupture of IAs [56]. Yamaguchi et al. proposed that estrogen deficiency leads to decreased expression of ERα and Sirt1, resulting in reduced levels of the Sirt1 protein, which promotes the activation of NLRP3, thereby inducing the accumulation of IL-1β and MMP-9, potentially leading to the rupture of IA. Their research findings revealed an increase in IL-1β and MMP-9 protein levels in the rat IA rupture model, supporting the notion that IA rupture is at least partially associated with the activation of the NOD-, LRR-, and pyrin-domain-containing protein 3 (NLRP3) after ERα depletion in the rat IA model [93]. Yuan et al. found that CXC chemokine receptor 3 (CXCR3) is a direct target of miR-34a, which modulates the phenotypic transformation of VSMCs in IAs by directly targeting CXCR3. CXCR3 modulates the expression of MMP-2 through ceRNA regulation by sharing overlapping miR-34a binding sites [72].

8. Conclusions

The formation, development, and rupture of IAs are complex processes influenced by both genetic and environmental factors. Despite significant research, the underlying mechanisms remain incompletely understood. However, the exploration of genetic and molecular markers associated with IA offers new avenues for identifying high-risk populations. Numerous studies have shown a strong correlation of the levels of inflammatory markers like MCP-1, IL-1β, and TNF-α with different stages of IA development. Similarly, oxidative stress proteins such as COX-2, NOX, and NOS are believed to play critical regulatory roles in IA pathogenesis.
Moving forward, large-scale, multicenter studies are needed to validate the sensitivity and specificity of these biomarkers, particularly across diverse racial populations, as previous studies may have been limited by sample size. Additionally, the development of non-invasive and cost-effective screening tools holds promise for predicting IA formation, growth, and rupture risk, ultimately facilitating early intervention and improved patient outcomes.

Author Contributions

Conceptualization, X.G. and Y.H.; methodology, E.Z., X.Y., H.S. and M.Z.; writing—original draft preparation, E.Z.; writing—review and editing, Y.H. and X.G.; project administration, Y.H.; funding acquisition, X.G. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from Ningbo Natural Science Foundation (2023J019) and the Ningbo Top Medical and Health Research Program (2022020304).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AAAbdominal aortic aneurysm
BCL2B-cell lymphoma-2
CASP1/ICECaspase 1
CD14Cluster of differentiation 14
CNSCentral nervous system
COL1A1Collagen type I alpha 1 chain
COL1A2Collagen type I alpha 2 chain
COL3A1Collagen type III alpha 1 chain
COL5A1Collagen type V alpha 1 chain
COL5A2Collagen type V alpha 2 chain
COX-1Cyclooxygenase-1
COX-2Cyclooxygenase-2
CSFCerebrospinal fluid
CTAComputed tomography angiography
CXCL12Chemokine (C-X-C motif) ligand 12
CXCR3CXC chemokine receptor 3
DNMT3ADNA methyltransferase 3 alpha
DPP-4Dipeptidyl peptidase-4
DSADigital subtraction angiography
ECMExtracellular matrix
ECsEndothelial cells
EDNRAEndothelin receptor type A
ELISAEnzyme-linked immunosorbent assay
eNOSEndothelial nitric oxide synthase
ET-1Endothelin-1
EVsExtracellular vesicles
FASFatty acid synthase
FTOFat mass and obesity-associated protein
GEOGene expression omnibus
gp130Glycoprotein 130
GREM1Gremlin 1
GSTA4Glutathione S-transferase alpha 4
GWASGenome-wide association study
H2O2Hydrogen peroxide
HBVSMCsHuman brain vascular smooth muscle cells
HDAC9Histone deacetylase 9
IAIntracranial aneurysm
IAsIntracranial aneurysms
IELInternal elastic lamina
IFN-γInterferon-gamma
IGF2BP1Insulin-like growth factor 2 mRNA binding protein 1
IGF2BP3Insulin-like growth factor 2 mRNA binding protein 3
IL-1βInterleukin-1β
IL-6Interleukin-6
IL-8Interleukin-8
iNOSInducible NOS
KLF5Kruppel-like factor 5
LOXLipoxygenase
KDM1ALysine-specific demethylase 1
LRPPRCLeucine-rich pentatricopeptide repeat containing
m1AN1-methyladenosine
m5C2′-O-methylation, 5-methylcytosine
m6AN6-methyladenosine
MAP3K10Mitogen-activated protein kinase 10
MBD2Methyl-CpG-binding domain protein 2
MCL-1Myeloid leukemia 1
MCP-1Monocyte chemoattractant protein-1
MethylMethylation
MIRA-seqMethylated-pyrosequencing
MMP-2Matrix metalloproteinase-2
MMP-3Matrix metalloproteinase-3
MMP-9Matrix metalloproteinase-9
MMP-13Matrix metalloproteinase-13
MMPsMatrix metalloproteinases
MRAMagnetic resonance angiography
NADPHNicotinamide Adenine Dinucleotide Phosphate
NFATC1Nuclear factor of activated T cell
NF-κBNuclear factor kappa-B
NLRP3The NOD-, LRR-, and pyrin domain-containing protein 3
nNOSNeuronal NOS
NONitric oxide
NOSNitric oxide synthase
NOXNicotinamide Adenine Dinucleotide Phosphate Oxidase
O2•-Superoxide
ONOO•-Peroxynitrite
PCAPrincipal component analyzed
PGE2Prostaglandin E2
PGG2Prostaglandin G2
PMNPolymorphonuclear neutrophils
PNPLA6Patatin-like phospholipase domain-containing protein 6
PTENPhosphatase and tensin homolog
PTGS2/COX2Cyclooxygenase-2
qRT-PCRQuantitative real-time polymerase chain reaction
RBM15RNA-binding motif protein 15
RIARuptured intracranial aneurysm
RNA-seqRNA sequencing
RNSReactive nitrogen species
ROSReactive oxygen species
SAHSubarachnoid hemorrhage
SERPINA3Serine protease inhibitor clade A member 3
SMCSmooth muscle cell
SMCsSmooth muscle cells
SNPsSingle nucleotide polymorphisms
SOCS3Cytokine signaling suppressor 3
TAMTumor-associated macrophage
TIMP4Tissue inhibitor of metalloproteinase 4
TNCTenascin C
TNF-αTumor necrosis factor-alpha
TRPC6Transient receptor potential-6
UIAUnruptured intracranial aneurysm
VCAM-1Vascular Cell Adhesion Molecule 1
VSMCVascular smooth muscle cell
WBWestern blotting
WSSWall shear stress
YTHDC1YT521-B homology domain-containing protein 1
YTHDF2YT521-B homology domain family member 2
YTHDF3YT521-B homology domain family member 2
ZNF217Zinc finger protein 217

References

  1. Brown, R.D., Jr.; Broderick, J.P. Unruptured intracranial aneurysms: Epidemiology, natural history, management options, and familial screening. Lancet Neurol. 2014, 13, 393–404. [Google Scholar] [CrossRef] [PubMed]
  2. Chalouhi, N.; Hoh, B.L.; Hasan, D. Review of cerebral aneurysm formation, growth, and rupture. Stroke 2013, 44, 3613–3622. [Google Scholar] [CrossRef] [PubMed]
  3. Li, M.H.; Chen, S.W.; Li, Y.D.; Chen, Y.C.; Cheng, Y.S.; Hu, D.J.; Tan, H.Q.; Wu, Q.; Wang, W.; Sun, Z.K.; et al. Prevalence of unruptured cerebral aneurysms in Chinese adults aged 35 to 75 years: A cross-sectional study. Ann. Intern. Med. 2013, 159, 514–521. [Google Scholar] [CrossRef]
  4. Vernooij, M.W.; Ikram, M.A.; Tanghe, H.L.; Vincent, A.J.; Hofman, A.; Krestin, G.P.; Niessen, W.J.; Breteler, M.M.; van der Lugt, A. Incidental findings on brain MRI in the general population. N. Engl. J. Med. 2007, 357, 1821–1828. [Google Scholar] [CrossRef]
  5. Chason, J.L.; Hindman, W.M. Berry aneurysms of the circle of Willis; results of a planned autopsy study. Neurology 1958, 8, 41–44. [Google Scholar] [CrossRef]
  6. Inagawa, T.; Hirano, A. Autopsy study of unruptured incidental intracranial aneurysms. Surg. Neurol. 1990, 34, 361–365. [Google Scholar] [CrossRef]
  7. Rackauskaite, D.; Svanborg, E.; Andersson, E.; Löwhagen, K.; Csajbok, L.; Nellgård, B. Prospective study: Long-term outcome at 12–15 years after aneurysmal subarachnoid hemorrhage. Acta Neurol. Scand. 2018, 138, 400–407. [Google Scholar] [CrossRef]
  8. Etminan, N.; Rinkel, G.J. Unruptured intracranial aneurysms: Development, rupture and preventive management. Nat. Rev. Neurol. 2016, 12, 699–713. [Google Scholar] [CrossRef]
  9. Etminan, N.; Dreier, R.; Buchholz, B.A.; Beseoglu, K.; Bruckner, P.; Matzenauer, C.; Torner, J.C.; Brown, R.D., Jr.; Steiger, H.J.; Hänggi, D.; et al. Age of collagen in intracranial saccular aneurysms. Stroke 2014, 45, 1757–1763. [Google Scholar] [CrossRef]
  10. Sluijter, J.P.; Smeets, M.B.; Velema, E.; Pasterkamp, G.; de Kleijn, D.P. Increased collagen turnover is only partly associated with collagen fiber deposition in the arterial response to injury. Cardiovasc. Res. 2004, 61, 186–195. [Google Scholar] [CrossRef]
  11. Rowe, A.J.; Finlay, H.M.; Canham, P.B. Collagen biomechanics in cerebral arteries and bifurcations assessed by polarizing microscopy. J. Vasc. Res. 2003, 40, 406–415. [Google Scholar] [CrossRef] [PubMed]
  12. Meng, H.; Tutino, V.M.; Xiang, J.; Siddiqui, A. High WSS or low WSS? Complex interactions of hemodynamics with intracranial aneurysm initiation, growth, and rupture: Toward a unifying hypothesis. AJNR Am. J. Neuroradiol. 2014, 35, 1254–1262. [Google Scholar] [CrossRef] [PubMed]
  13. Frösen, J.; Tulamo, R.; Paetau, A.; Laaksamo, E.; Korja, M.; Laakso, A.; Niemelä, M.; Hernesniemi, J. Saccular intracranial aneurysm: Pathology and mechanisms. Acta Neuropathol. 2012, 123, 773–786. [Google Scholar] [CrossRef] [PubMed]
  14. Pradilla, G.; Wicks, R.T.; Hadelsberg, U.; Gailloud, P.; Coon, A.L.; Huang, J.; Tamargo, R.J. Accuracy of computed tomography angiography in the diagnosis of intracranial aneurysms. World Neurosurg. 2013, 80, 845–852. [Google Scholar] [CrossRef]
  15. Sailer, A.M.; Wagemans, B.A.; Nelemans, P.J.; de Graaf, R.; van Zwam, W.H. Diagnosing intracranial aneurysms with MR angiography: Systematic review and meta-analysis. Stroke 2014, 45, 119–126. [Google Scholar] [CrossRef]
  16. Etminan, N.; Buchholz, B.A.; Dreier, R.; Bruckner, P.; Torner, J.C.; Steiger, H.J.; Hänggi, D.; Macdonald, R.L. Cerebral aneurysms: Formation, progression, and developmental chronology. Transl. Stroke Res. 2014, 5, 167–173. [Google Scholar] [CrossRef]
  17. Alg, V.S.; Sofat, R.; Houlden, H.; Werring, D.J. Genetic risk factors for intracranial aneurysms: A meta-analysis in more than 116,000 individuals. Neurology 2013, 80, 2154–2165. [Google Scholar] [CrossRef]
  18. Korja, M.; Silventoinen, K.; McCarron, P.; Zdravkovic, S.; Skytthe, A.; Haapanen, A.; de Faire, U.; Pedersen, N.L.; Christensen, K.; Koskenvuo, M.; et al. Genetic epidemiology of spontaneous subarachnoid hemorrhage: Nordic Twin Study. Stroke 2010, 41, 2458–2462. [Google Scholar] [CrossRef]
  19. Vlak, M.H.; Rinkel, G.J.; Greebe, P.; Algra, A. Independent risk factors for intracranial aneurysms and their joint effect: A case-control study. Stroke 2013, 44, 984–987. [Google Scholar] [CrossRef]
  20. Meng, H.; Wang, Z.; Hoi, Y.; Gao, L.; Metaxa, E.; Swartz, D.D.; Kolega, J. Complex hemodynamics at the apex of an arterial bifurcation induces vascular remodeling resembling cerebral aneurysm initiation. Stroke 2007, 38, 1924–1931. [Google Scholar] [CrossRef]
  21. Miura, Y.; Ishida, F.; Umeda, Y.; Tanemura, H.; Suzuki, H.; Matsushima, S.; Shimosaka, S.; Taki, W. Low wall shear stress is independently associated with the rupture status of middle cerebral artery aneurysms. Stroke 2013, 44, 519–521. [Google Scholar] [CrossRef] [PubMed]
  22. Hasan, D.; Chalouhi, N.; Jabbour, P.; Hashimoto, T. Macrophage imbalance (M1 vs. M2) and upregulation of mast cells in wall of ruptured human cerebral aneurysms: Preliminary results. J. Neuroinflammation 2012, 9, 222. [Google Scholar] [CrossRef] [PubMed]
  23. Sibon, I.; Mercier, N.; Darret, D.; Lacolley, P.; Lamazière, J.M. Association between semicarbazide-sensitive amine oxidase, a regulator of the glucose transporter, and elastic lamellae thinning during experimental cerebral aneurysm development: Laboratory investigation. J. Neurosurg. 2008, 108, 558–566. [Google Scholar] [CrossRef] [PubMed]
  24. Strimbu, K.; Tavel, J.A. What are biomarkers? Curr. Opin. HIV AIDS 2010, 5, 463–466. [Google Scholar] [CrossRef]
  25. Wang, W.H.; Wang, Y.H.; Zheng, L.L.; Li, X.W.; Hao, F.; Guo, D. MicroRNA-29a: A potential biomarker in the development of intracranial aneurysm. J. Neurol. Sci. 2016, 364, 84–89. [Google Scholar] [CrossRef]
  26. Kao, H.W.; Lee, K.W.; Kuo, C.L.; Huang, C.S.; Tseng, W.M.; Liu, C.S.; Lin, C.P. Interleukin-6 as a Prognostic Biomarker in Ruptured Intracranial Aneurysms. PLoS ONE 2015, 10, e0132115. [Google Scholar] [CrossRef]
  27. Richards, E.J. Inherited epigenetic variation--revisiting soft inheritance. Nat. Rev. Genet. 2006, 7, 395–401. [Google Scholar] [CrossRef]
  28. Pritchard, C.C.; Cheng, H.H.; Tewari, M. MicroRNA profiling: Approaches and considerations. Nat. Rev. Genet. 2012, 13, 358–369. [Google Scholar] [CrossRef]
  29. Hosaka, K.; Hoh, B.L. Inflammation and cerebral aneurysms. Transl. Stroke Res. 2014, 5, 190–198. [Google Scholar] [CrossRef]
  30. Liu, Q.; Li, K.; He, H.; Miao, Z.; Cui, H.; Wu, J.; Ding, S.; Wen, Z.; Chen, J.; Lu, X.; et al. The markers and risk stratification model of intracranial aneurysm instability in a large Chinese cohort. Sci. Bull. 2023, 68, 1162–1175. [Google Scholar] [CrossRef]
  31. Tutino, V.M.; Poppenberg, K.E.; Li, L.; Shallwani, H.; Jiang, K.; Jarvis, J.N.; Sun, Y.; Snyder, K.V.; Levy, E.I.; Siddiqui, A.H.; et al. Biomarkers from circulating neutrophil transcriptomes have potential to detect unruptured intracranial aneurysms. J. Transl. Med. 2018, 16, 373. [Google Scholar] [CrossRef] [PubMed]
  32. Kamińska, J.; Maciejczyk, M.; Ćwiklińska, A.; Matowicka-Karna, J.; Koper-Lenkiewicz, O.M. Pro-Inflammatory and Anti-Inflammatory Cytokines Levels are Significantly Altered in Cerebrospinal Fluid of Unruptured Intracranial Aneurysm (UIA) Patients. J. Inflamm. Res. 2022, 15, 6245–6261. [Google Scholar] [CrossRef] [PubMed]
  33. Luo, L.; Ma, X.; Kong, D.; Dai, Y.; Li, T.; Yu, H.; Liu, J.; Li, M.; Xu, Y.; Xiang, G.; et al. Multiomics integrated analysis and experimental validation identify TLR4 and ALOX5 as oxidative stress-related biomarkers in intracranial aneurysms. J. Neuroinflammation 2024, 21, 225. [Google Scholar] [CrossRef] [PubMed]
  34. Kadry, H.; Noorani, B.; Cucullo, L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  35. Gaetani, L.; Blennow, K.; Calabresi, P.; Di Filippo, M.; Parnetti, L.; Zetterberg, H. Neurofilament light chain as a biomarker in neurological disorders. J. Neurol. Neurosurg. Psychiatry 2019, 90, 870–881. [Google Scholar] [CrossRef]
  36. Wiśniewski, K.; Zaczkowski, K.; Szmyd, B.M.; Popęda, M.; Bieńkowski, M.; Posmyk, B.; Bobeff, E.J.; Jaskólski, D.J. Evaluation of CSF 8-iso-prostaglandin F2α and erythrocyte anisocytosis as prognostic biomarkers for delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Sci. Rep. 2024, 14, 11302. [Google Scholar] [CrossRef]
  37. Roder, C.; Kasuya, H.; Harati, A.; Tatagiba, M.; Inoue, I.; Krischek, B. Meta-analysis of microarray gene expression studies on intracranial aneurysms. Neuroscience 2012, 201, 105–113. [Google Scholar] [CrossRef]
  38. McColgan, P.; Thant, K.Z.; Sharma, P. The genetics of sporadic ruptured and unruptured intracranial aneurysms: A genetic meta-analysis of 8 genes and 13 polymorphisms in approximately 20,000 individuals. J. Neurosurg. 2010, 112, 714–721. [Google Scholar] [CrossRef]
  39. Slowik, A.; Borratynska, A.; Turaj, W.; Pera, J.; Dziedzic, T.; Figlewicz, D.A.; Betlej, M.; Krzyszkowski, T.; Czepko, R.; Szczudlik, A. Alpha1-antichymotrypsin gene (SERPINA3) A/T polymorphism as a risk factor for aneurysmal subarachnoid hemorrhage. Stroke 2005, 36, 737–740. [Google Scholar] [CrossRef]
  40. Liu, W.; Zhu, Y.; Ge, M.; Pang, Q.; Yu, Y. Polymorphism rs4934 of SERPINA3 and sporadic intracranial aneurysms in the Chinese population. Cerebrovasc. Dis. 2010, 29, 68–72. [Google Scholar] [CrossRef]
  41. Bakker, M.K.; Ruigrok, Y.M. Genetics of Intracranial Aneurysms. Stroke 2021, 52, 3004–3012. [Google Scholar] [CrossRef]
  42. Yasuno, K.; Bakırcıoğlu, M.; Low, S.K.; Bilgüvar, K.; Gaál, E.; Ruigrok, Y.M.; Niemelä, M.; Hata, A.; Bijlenga, P.; Kasuya, H.; et al. Common variant near the endothelin receptor type A (EDNRA) gene is associated with intracranial aneurysm risk. Proc. Natl. Acad. Sci. USA 2011, 108, 19707–19712. [Google Scholar] [CrossRef]
  43. Yu, L.; Wang, J.; Wang, S.; Zhang, D.; Zhao, Y.; Wang, R.; Zhao, J. DNA Methylation Regulates Gene Expression in Intracranial Aneurysms. World Neurosurg. 2017, 105, 28–36. [Google Scholar] [CrossRef]
  44. Ma, J.; Joehanes, R.; Liu, C.; Keshawarz, A.; Hwang, S.J.; Bui, H.; Tejada, B.; Sooda, M.; Munson, P.J.; Demirkale, C.Y.; et al. Elucidating the genetic architecture of DNA methylation to identify promising molecular mechanisms of disease. Sci. Rep. 2022, 12, 19564. [Google Scholar] [CrossRef]
  45. Zhao, J.; Xu, T.; Zhou, S.; Zhang, J.; Wu, Y.; Zhou, C.; Sun, J.; Gao, X.; Huang, Y. DNA methylation of the MAP3K10 gene may participate in the development of intracranial aneurysm. Gene 2023, 851, 147024. [Google Scholar] [CrossRef]
  46. Xu, T.; Yu, X.; Zhou, S.; Wu, Y.; Deng, X.; Wu, Y.; Wang, S.; Gao, X.; Nie, S.; Zhou, C.; et al. DNA methylation and mRNA expression of glutathione S-transferase alpha 4 are associated with intracranial aneurysms in a gender-dependent manner. Front. Genet. 2022, 13, 1079455. [Google Scholar] [CrossRef]
  47. Zhou, S.; Zhang, J.; Zhou, C.; Gong, F.; Zhu, X.; Pan, X.; Sun, J.; Gao, X.; Huang, Y. DNA Methylation of Patatin-Like Phospholipase Domain-Containing Protein 6 Gene Contributes to the Risk of Intracranial Aneurysm in Males. Front. Aging Neurosci. 2022, 14, 885680. [Google Scholar] [CrossRef]
  48. Maimaiti, A.; Turhon, M.; Abulaiti, A.; Dilixiati, Y.; Zhang, F.; Axieer, A.; Kadeer, K.; Zhang, Y.; Maimaitili, A.; Yang, X. DNA methylation regulator-mediated modification patterns and risk of intracranial aneurysm: A multi-omics and epigenome-wide association study integrating machine learning, Mendelian randomization, eQTL and mQTL data. J. Transl. Med. 2023, 21, 660. [Google Scholar] [CrossRef]
  49. Zhong, Z.; Wu, J.; Yuan, K.; Song, Z.; Ma, Z.; Zhong, Y.; Fang, X.; Zhang, W. Upregulation of microRNA-205 is a potential biomarker for intracranial aneurysms. Neuroreport 2019, 30, 812–816. [Google Scholar] [CrossRef]
  50. Feng, Z.; Zhang, X.; Li, L.; Wang, C.; Feng, M.; Zhao, K.; Zhao, R.; Liu, J.; Fang, Y. Tumor-associated macrophage-derived exosomal microRNA-155-5p stimulates intracranial aneurysm formation and macrophage infiltration. Clin. Sci. 2019, 133, 2265–2282. [Google Scholar] [CrossRef]
  51. Yu, G.; Liu, P.; Shi, Y.; Li, S.; Liu, Y.; Fan, Z.; Zhu, W. Stimulation of endothelial progenitor cells by microRNA-31a-5p to induce endothelialization in an aneurysm neck after coil embolization by modulating the Axin1-mediated β-catenin/vascular endothelial growth factor pathway. J. Neurosurg. 2019, 133, 918–926. [Google Scholar] [CrossRef]
  52. Xiong, W.; Yao, W.; Gao, Z.; Liu, K. Rs12976445 polymorphism is associated with the risk of post-SAH re-bleeding by modulating the expression of microRNA-125 and ET-1. Sci. Rep. 2022, 12, 2062. [Google Scholar] [CrossRef]
  53. Holcomb, M.; Ding, Y.H.; Dai, D.; McDonald, R.J.; McDonald, J.S.; Kallmes, D.F.; Kadirvel, R. RNA-Sequencing Analysis of Messenger RNA/MicroRNA in a Rabbit Aneurysm Model Identifies Pathways and Genes of Interest. AJNR Am. J. Neuroradiol. 2015, 36, 1710–1715. [Google Scholar] [CrossRef]
  54. Jiang, Y.; Zhang, M.; He, H.; Chen, J.; Zeng, H.; Li, J.; Duan, R. MicroRNA/mRNA profiling and regulatory network of intracranial aneurysm. BMC Med. Genom. 2013, 6, 36. [Google Scholar] [CrossRef]
  55. Zhao, W.; Zhang, H.; Su, J.Y. MicroRNA-29a contributes to intracranial aneurysm by regulating the mitochondrial apoptotic pathway. Mol. Med. Rep. 2018, 18, 2945–2954. [Google Scholar] [CrossRef]
  56. Zheng, Z.; Chen, Y.; Wang, Y.; Li, Y.; Cheng, Q. MicroRNA-513b-5p targets COL1A1 and COL1A2 associated with the formation and rupture of intracranial aneurysm. Sci. Rep. 2021, 11, 14897. [Google Scholar] [CrossRef]
  57. Fan, W.; Liu, Y.; Li, C.; Qu, X.; Zheng, G.; Zhang, Q.; Pan, Z.; Wang, Y.; Rong, J. microRNA-331-3p maintains the contractile type of vascular smooth muscle cells by regulating TNF-α and CD14 in intracranial aneurysm. Neuropharmacology 2020, 164, 107858. [Google Scholar] [CrossRef]
  58. Bekelis, K.; Kerley-Hamilton, J.S.; Teegarden, A.; Tomlinson, C.R.; Kuintzle, R.; Simmons, N.; Singer, R.J.; Roberts, D.W.; Kellis, M.; Hendrix, D.A. MicroRNA and gene expression changes in unruptured human cerebral aneurysms. J. Neurosurg. 2016, 125, 1390–1399. [Google Scholar] [CrossRef]
  59. Lv, C.; Wang, J.; Dai, S.; Chen, Y.; Jiang, X.; Li, X. Long non-coding RNA NORAD induces phenotypic regulation of vascular smooth muscle cells through regulating microRNA-136-5p-targeted KDM1A. Cell Cycle 2021, 20, 2137–2148. [Google Scholar] [CrossRef]
  60. Cai, Y.; Huang, D.; Ma, W.; Wang, M.; Qin, Q.; Jiang, Z.; Liu, M. Histone deacetylase 9 inhibition upregulates microRNA-92a to repress the progression of intracranial aneurysm via silencing Bcl-2-like protein 11. J. Drug Target. 2021, 29, 761–770. [Google Scholar] [CrossRef]
  61. Liu, D.; Han, L.; Wu, X.; Yang, X.; Zhang, Q.; Jiang, F. Genome-wide microRNA changes in human intracranial aneurysms. BMC Neurol. 2014, 14, 188. [Google Scholar] [CrossRef]
  62. Guo, D.; Wang, Y.W.; Yan, L.; Ma, J.; Han, X.W.; Shui, S.F. Dysregulation of microRNA-23b-3p contributes to the development of intracranial aneurysms by targeting phosphatase and tensin homolog. Int. J. Mol. Med. 2018, 42, 1637–1643. [Google Scholar] [CrossRef]
  63. Jin, H.; Li, C.; Ge, H.; Jiang, Y.; Li, Y. Circulating microRNA: A novel potential biomarker for early diagnosis of intracranial aneurysm rupture a case control study. J. Transl. Med. 2013, 11, 296. [Google Scholar] [CrossRef]
  64. Su, X.W.; Chan, A.H.; Lu, G.; Lin, M.; Sze, J.; Zhou, J.Y.; Poon, W.S.; Liu, Q.; Zheng, V.Z.; Wong, G.K. Circulating microRNA 132-3p and 324-3p Profiles in Patients after Acute Aneurysmal Subarachnoid Hemorrhage. PLoS ONE 2015, 10, e0144724. [Google Scholar] [CrossRef]
  65. Supriya, M.; Christopher, R.; Devi, B.I.; Bhat, D.I.; Shukla, D.; Kalpana, S.R. Altered MicroRNA Expression in Intracranial Aneurysmal Tissues: Possible Role in TGF-β Signaling Pathway. Cell Mol. Neurobiol. 2022, 42, 2393–2405. [Google Scholar] [CrossRef]
  66. Luo, J.; Jin, H.; Jiang, Y.; Ge, H.; Wang, J.; Li, Y. Aberrant Expression of microRNA-9 Contributes to Development of Intracranial Aneurysm by Suppressing Proliferation and Reducing Contractility of Smooth Muscle Cells. Med. Sci. Monit. 2016, 22, 4247–4253. [Google Scholar] [CrossRef]
  67. Yang, G.; Qin, H.; Liu, B.; Zhao, X.; Yin, H. Mesenchymal stem cells-derived exosomes modulate vascular endothelial injury via miR-144-5p/PTEN in intracranial aneurysm. Hum. Cell 2021, 34, 1346–1359. [Google Scholar] [CrossRef]
  68. Liao, B.; Zhou, M.X.; Zhou, F.K.; Luo, X.M.; Zhong, S.X.; Zhou, Y.F.; Qin, Y.S.; Li, P.P.; Qin, C. Exosome-Derived MiRNAs as Biomarkers of the Development and Progression of Intracranial Aneurysms. J. Atheroscler. Thromb. 2020, 27, 545–610. [Google Scholar] [CrossRef]
  69. Li, P.; Zhang, Q.; Wu, X.; Yang, X.; Zhang, Y.; Li, Y.; Jiang, F. Circulating microRNAs serve as novel biological markers for intracranial aneurysms. J. Am. Heart Assoc. 2014, 3, e000972. [Google Scholar] [CrossRef]
  70. Zou, L.; Hou, Y.; Yu, B.; Li, S.; Du, Y. The effect of intravascular interventional embolization and craniotomy on MMP-2, MMP-9 and caspase3 in serum of intracranial aneurysm patients. Exp. Ther. Med. 2018, 16, 4511–4518. [Google Scholar] [CrossRef]
  71. Xu, J.; Yan, S.; Tan, H.; Ma, L.; Feng, H.; Han, H.; Pan, M.; Yu, L.; Fang, C. The miR-143/145 cluster reverses the regulation effect of KLF5 in smooth muscle cells with proliferation and contractility in intracranial aneurysm. Gene 2018, 679, 266–273. [Google Scholar] [CrossRef]
  72. Yuan, X.; Bian, X.; Wei, W.; Bao, Q.; Liu, P.; Jiang, W. miR-34a regulates phenotypic modulation of vascular smooth muscle cells in intracranial aneurysm by targeting CXCR3 and MMP-2. Genet. Mol. Biol. 2021, 44, e20200124. [Google Scholar] [CrossRef]
  73. Marks, L.B.; Yu, X.; Vujaskovic, Z.; Small, W., Jr.; Folz, R.; Anscher, M.S. Radiation-induced lung injury. Semin. Radiat. Oncol. 2003, 13, 333–345. [Google Scholar] [CrossRef]
  74. Skoczylas, J.Z.; Bentzen, S.M.; Overgaard, M.; Overgaard, J. Time course of radiological lung density changes after postmastectomy radiotherapy. Acta Oncol. 2000, 39, 181–187. [Google Scholar] [CrossRef]
  75. Ozsolak, F.; Milos, P.M. RNA sequencing: Advances, challenges and opportunities. Nat. Rev. Genet. 2011, 12, 87–98. [Google Scholar] [CrossRef]
  76. Lin, W.; Ye, H.; You, K.; Chen, L. Up-regulation of circ_LARP4 suppresses cell proliferation and migration in ovarian cancer by regulating miR-513b-5p/LARP4 axis. Cancer Cell Int. 2020, 20, 5. [Google Scholar] [CrossRef]
  77. Desrosiers, R.; Friderici, K.; Rottman, F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc. Natl. Acad. Sci. USA 1974, 71, 3971–3975. [Google Scholar] [CrossRef]
  78. Zhang, B.; Wu, Q.; Li, B.; Wang, D.; Wang, L.; Zhou, Y.L. m(6)A regulator-mediated methylation modification patterns and tumor microenvironment infiltration characterization in gastric cancer. Mol. Cancer 2020, 19, 53. [Google Scholar] [CrossRef]
  79. Zhang, H.; Zhao, L.; Li, S.; Wang, J.; Feng, C.; Li, T.; Du, X. N6-Methylandenosine-Related lncRNAs in Tumor Microenvironment Are Potential Prognostic Biomarkers in Colon Cancer. Front. Oncol. 2021, 11, 697949. [Google Scholar] [CrossRef]
  80. Maimaiti, A.; Turhon, M.; Cheng, X.; Su, R.; Kadeer, K.; Axier, A.; Ailaiti, D.; Aili, Y.; Abudusalamu, R.; Kuerban, A.; et al. m6A regulator-mediated RNA methylation modification patterns and immune microenvironment infiltration characterization in patients with intracranial aneurysms. Front. Neurol. 2022, 13, 889141. [Google Scholar] [CrossRef]
  81. Li, S.; Zhang, Q.; Weng, L.; Han, Y.; Li, J. Novel insight into m6A regulator-mediated methylation modification patterns and immune characteristics in intracranial aneurysm. Front. Aging Neurosci. 2022, 14, 973258. [Google Scholar] [CrossRef]
  82. Zhang, H.F.; Zhao, M.G.; Liang, G.B.; Song, Z.Q.; Li, Z.Q. Expression of pro-inflammatory cytokines and the risk of intracranial aneurysm. Inflammation 2013, 36, 1195–1200. [Google Scholar] [CrossRef]
  83. Aoki, T.; Kataoka, H.; Ishibashi, R.; Nozaki, K.; Egashira, K.; Hashimoto, N. Impact of monocyte chemoattractant protein-1 deficiency on cerebral aneurysm formation. Stroke 2009, 40, 942–951. [Google Scholar] [CrossRef]
  84. Chu, C.; Xu, G.; Li, X.; Duan, Z.; Tao, L.; Cai, H.; Yang, M.; Zhang, X.; Chen, B.; Zheng, Y.; et al. Sustained expression of MCP-1 induced low wall shear stress loading in conjunction with turbulent flow on endothelial cells of intracranial aneurysm. J. Cell Mol. Med. 2021, 25, 110–119. [Google Scholar] [CrossRef]
  85. Lai, X.L.; Deng, Z.F.; Zhu, X.G.; Chen, Z.H. Apc gene suppresses intracranial aneurysm formation and rupture through inhibiting the NF-κB signaling pathway mediated inflammatory response. Biosci. Rep. 2019, 39, BSR20181909. [Google Scholar] [CrossRef]
  86. Liu, Y.; Zhang, Y.; Dai, D.; Xu, Z. Expression of NF-κB, MCP-1 and MMP-9 in a Cerebral Aneurysm Rabbit Model. Can. J. Neurol. Sci. 2014, 41, 200–205. [Google Scholar] [CrossRef]
  87. Ait-Oufella, H.; Taleb, S.; Mallat, Z.; Tedgui, A. Recent advances on the role of cytokines in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 969–979. [Google Scholar] [CrossRef]
  88. Ali, M.S.; Starke, R.M.; Jabbour, P.M.; Tjoumakaris, S.I.; Gonzalez, L.F.; Rosenwasser, R.H.; Owens, G.K.; Koch, W.J.; Greig, N.H.; Dumont, A.S. TNF-α induces phenotypic modulation in cerebral vascular smooth muscle cells: Implications for cerebral aneurysm pathology. J. Cereb. Blood Flow. Metab. 2013, 33, 1564–1573. [Google Scholar] [CrossRef]
  89. Starke, R.M.; Raper, D.M.; Ding, D.; Chalouhi, N.; Owens, G.K.; Hasan, D.M.; Medel, R.; Dumont, A.S. Tumor necrosis factor-α modulates cerebral aneurysm formation and rupture. Transl. Stroke Res. 2014, 5, 269–277. [Google Scholar] [CrossRef]
  90. Kanematsu, Y.; Kanematsu, M.; Kurihara, C.; Tada, Y.; Tsou, T.L.; van Rooijen, N.; Lawton, M.T.; Young, W.L.; Liang, E.I.; Nuki, Y.; et al. Critical roles of macrophages in the formation of intracranial aneurysm. Stroke 2011, 42, 173–178. [Google Scholar] [CrossRef]
  91. Starke, R.M.; Chalouhi, N.; Jabbour, P.M.; Tjoumakaris, S.I.; Gonzalez, L.F.; Rosenwasser, R.H.; Wada, K.; Shimada, K.; Hasan, D.M.; Greig, N.H.; et al. Critical role of TNF-α in cerebral aneurysm formation and progression to rupture. J. Neuroinflammation 2014, 11, 77. [Google Scholar] [CrossRef] [PubMed]
  92. Jayaraman, T.; Berenstein, V.; Li, X.; Mayer, J.; Silane, M.; Shin, Y.S.; Niimi, Y.; Kiliç, T.; Gunel, M.; Berenstein, A. Tumor necrosis factor alpha is a key modulator of inflammation in cerebral aneurysms. Neurosurgery 2005, 57, 558–564; discussion 558–564. [Google Scholar] [CrossRef] [PubMed]
  93. Yamaguchi, T.; Miyamoto, T.; Shikata, E.; Yamaguchi, I.; Shimada, K.; Yagi, K.; Tada, Y.; Korai, M.; Kitazato, K.T.; Kanematsu, Y.; et al. Activation of the NLRP3/IL-1β/MMP-9 pathway and intracranial aneurysm rupture associated with the depletion of ERα and Sirt1 in oophorectomized rats. J. Neurosurg. 2023, 138, 191–198. [Google Scholar] [CrossRef]
  94. Kamińska, J.; Lyson, T.; Chrzanowski, R.; Sawicki, K.; Milewska, A.J.; Tylicka, M.; Zińczuk, J.; Matowicka-Karna, J.; Dymicka-Piekarska, V.; Mariak, Z.; et al. Ratio of IL-8 in CSF versus Serum Is Elevated in Patients with Unruptured Brain Aneurysm. J. Clin. Med. 2020, 9, 1761. [Google Scholar] [CrossRef]
  95. Kamińska, J.; Tylicka, M.; Dymicka-Piekarska, V.; Mariak, Z.; Matowicka-Karna, J.; Koper-Lenkiewicz, O.M. Canonical NF-κB signaling pathway and GRO-α/CXCR2 axis are activated in unruptured intracranial aneurysm patients. Sci. Rep. 2022, 12, 21375. [Google Scholar] [CrossRef]
  96. Aoki, T.; Nishimura, M.; Matsuoka, T.; Yamamoto, K.; Furuyashiki, T.; Kataoka, H.; Kitaoka, S.; Ishibashi, R.; Ishibazawa, A.; Miyamoto, S.; et al. PGE(2)-EP(2) signalling in endothelium is activated by haemodynamic stress and induces cerebral aneurysm through an amplifying loop via NF-κB. Br. J. Pharmacol. 2011, 163, 1237–1249. [Google Scholar] [CrossRef]
  97. Sun, Z.H.; Liu, F.; Kong, L.L.; Ji, P.M.; Huang, L.; Zhou, H.M.; Sun, R.; Luo, J.; Li, W.Z. Interruption of TRPC6-NFATC1 signaling inhibits NADPH oxidase 4 and VSMCs phenotypic switch in intracranial aneurysm. Biomed. Pharmacother. 2023, 161, 114480. [Google Scholar] [CrossRef]
  98. Aoki, T.; Kataoka, H.; Ishibashi, R.; Nozaki, K.; Morishita, R.; Hashimoto, N. Reduced collagen biosynthesis is the hallmark of cerebral aneurysm: Contribution of interleukin-1beta and nuclear factor-kappaB. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1080–1086. [Google Scholar] [CrossRef]
  99. Sprague, A.H.; Khalil, R.A. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem. Pharmacol. 2009, 78, 539–552. [Google Scholar] [CrossRef]
  100. Petrasek, J.; Bala, S.; Csak, T.; Lippai, D.; Kodys, K.; Menashy, V.; Barrieau, M.; Min, S.Y.; Kurt-Jones, E.A.; Szabo, G. IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J. Clin. Investig. 2012, 122, 3476–3489. [Google Scholar] [CrossRef]
  101. Johnston, W.F.; Salmon, M.; Su, G.; Lu, G.; Stone, M.L.; Zhao, Y.; Owens, G.K.; Upchurch, G.R., Jr.; Ailawadi, G. Genetic and pharmacologic disruption of interleukin-1β signaling inhibits experimental aortic aneurysm formation. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 294–304. [Google Scholar] [CrossRef] [PubMed]
  102. Moriwaki, T.; Takagi, Y.; Sadamasa, N.; Aoki, T.; Nozaki, K.; Hashimoto, N. Impaired progression of cerebral aneurysms in interleukin-1beta-deficient mice. Stroke 2006, 37, 900–905. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, Q.; Zhang, Y.; Zhu, C.; Liu, W.; Ma, X.; Chen, J.; Mo, S.; Dong, L.; Wang, N.; Wu, J.; et al. Serum IL-1, Pyroptosis and Intracranial Aneurysm Wall Enhancement: Analysis Integrating Radiology, Serum Cytokines and Histology. Front. Cardiovasc. Med. 2022, 9, 818789. [Google Scholar] [CrossRef]
  104. Sun, S.C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef]
  105. Aoki, T.; Kataoka, H.; Shimamura, M.; Nakagami, H.; Wakayama, K.; Moriwaki, T.; Ishibashi, R.; Nozaki, K.; Morishita, R.; Hashimoto, N. NF-kappaB is a key mediator of cerebral aneurysm formation. Circulation 2007, 116, 2830–2840. [Google Scholar] [CrossRef]
  106. Zhou, H.; Khan, D.; Hussain, S.M.; Gerdes, N.; Hagenbeck, C.; Rana, M.; Cornelius, J.F.; Muhammad, S. Colchicine prevents oxidative stress-induced endothelial cell senescence via blocking NF-κB and MAPKs: Implications in vascular diseases. J. Inflamm. 2023, 20, 41. [Google Scholar] [CrossRef]
  107. Nakayama, K. cAMP-response element-binding protein (CREB) and NF-κB transcription factors are activated during prolonged hypoxia and cooperatively regulate the induction of matrix metalloproteinase MMP1. J. Biol. Chem. 2013, 288, 22584–22595. [Google Scholar] [CrossRef]
  108. Zhou, H.; Zhao, K.; Li, W.; Yang, N.; Liu, Y.; Chen, C.; Wei, T. The interactions between pristine graphene and macrophages and the production of cytokines/chemokines via TLR- and NF-κB-related signaling pathways. Biomaterials 2012, 33, 6933–6942. [Google Scholar] [CrossRef]
  109. Chucair-Elliott, A.J.; Conrady, C.; Zheng, M.; Kroll, C.M.; Lane, T.E.; Carr, D.J. Microglia-induced IL-6 protects against neuronal loss following HSV-1 infection of neural progenitor cells. Glia 2014, 62, 1418–1434. [Google Scholar] [CrossRef]
  110. Taga, T.; Hibi, M.; Hirata, Y.; Yamasaki, K.; Yasukawa, K.; Matsuda, T.; Hirano, T.; Kishimoto, T. Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell 1989, 58, 573–581. [Google Scholar] [CrossRef]
  111. Monsour, M.; Croci, D.M.; Grüter, B.E.; Taussky, P.; Marbacher, S.; Agazzi, S. Cerebral Aneurysm and Interleukin-6: A Key Player in Aneurysm Generation and Rupture or Just One of the Multiple Factors? Transl. Stroke Res. 2023, 14, 631–639. [Google Scholar] [CrossRef] [PubMed]
  112. Hodes, G.E.; Ménard, C.; Russo, S.J. Integrating Interleukin-6 into depression diagnosis and treatment. Neurobiol. Stress. 2016, 4, 15–22. [Google Scholar] [CrossRef] [PubMed]
  113. Luo, Y.; Zheng, S.G. Hall of Fame among Pro-inflammatory Cytokines: Interleukin-6 Gene and Its Transcriptional Regulation Mechanisms. Front. Immunol. 2016, 7, 604. [Google Scholar] [CrossRef]
  114. Ikedo, T.; Minami, M.; Kataoka, H.; Hayashi, K.; Nagata, M.; Fujikawa, R.; Higuchi, S.; Yasui, M.; Aoki, T.; Fukuda, M.; et al. Dipeptidyl Peptidase-4 Inhibitor Anagliptin Prevents Intracranial Aneurysm Growth by Suppressing Macrophage Infiltration and Activation. J. Am. Heart Assoc. 2017, 6, e004777. [Google Scholar] [CrossRef]
  115. Wajima, D.; Hourani, S.; Dodd, W.; Patel, D.; Jones, C.; Motwani, K.; Fazal, H.Z.; Hosaka, K.; Hoh, B.L. Interleukin-6 Promotes Murine Estrogen Deficiency-Associated Cerebral Aneurysm Rupture. Neurosurgery 2020, 86, 583–592. [Google Scholar] [CrossRef]
  116. Kamińska, J.; Dymicka-Piekarska, V.; Chrzanowski, R.; Sawicki, K.; Milewska, A.J.; Zińczuk, J.; Tylicka, M.; Jadeszko, M.; Mariak, Z.; Kratz, E.M.; et al. IL-6 Quotient (The Ratio of Cerebrospinal Fluid IL-6 to Serum IL-6) as a Biomarker of an Unruptured Intracranial Aneurysm. J. Inflamm. Res. 2021, 14, 6103–6114. [Google Scholar] [CrossRef]
  117. Croci, D.M.; Wanderer, S.; Strange, F.; Grüter, B.E.; Sivanrupan, S.; Andereggen, L.; Casoni, D.; von Gunten, M.; Widmer, H.R.; Di Santo, S.; et al. Tocilizumab Reduces Vasospasms, Neuronal Cell Death, and Microclot Formation in a Rabbit Model of Subarachnoid Hemorrhage. Transl. Stroke Res. 2021, 12, 894–904. [Google Scholar] [CrossRef]
  118. Griendling, K.K.; FitzGerald, G.A. Oxidative stress and cardiovascular injury: Part I: Basic mechanisms and in vivo monitoring of ROS. Circulation 2003, 108, 1912–1916. [Google Scholar] [CrossRef]
  119. Starke, R.M.; Chalouhi, N.; Ali, M.S.; Jabbour, P.M.; Tjoumakaris, S.I.; Gonzalez, L.F.; Rosenwasser, R.H.; Koch, W.J.; Dumont, A.S. The role of oxidative stress in cerebral aneurysm formation and rupture. Curr. Neurovasc. Res. 2013, 10, 247–255. [Google Scholar] [CrossRef]
  120. Chalouhi, N.; Chitale, R.; Jabbour, P.; Tjoumakaris, S.; Dumont, A.S.; Rosenwasser, R.; Gonzalez, L.F. The case for family screening for intracranial aneurysms. Neurosurg. Focus. 2011, 31, E8. [Google Scholar] [CrossRef]
  121. Talukder, M.A.; Johnson, W.M.; Varadharaj, S.; Lian, J.; Kearns, P.N.; El-Mahdy, M.A.; Liu, X.; Zweier, J.L. Chronic cigarette smoking causes hypertension, increased oxidative stress, impaired NO bioavailability, endothelial dysfunction, and cardiac remodeling in mice. Am. J. Physiol. Heart Circ. Physiol. 2011, 300, H388–H396. [Google Scholar] [CrossRef] [PubMed]
  122. Juvela, S. Prevalence of and risk factors for intracranial aneurysms. Lancet Neurol. 2011, 10, 595–597. [Google Scholar] [CrossRef] [PubMed]
  123. Montezano, A.C.; Touyz, R.M. Molecular mechanisms of hypertension--reactive oxygen species and antioxidants: A basic science update for the clinician. Can. J. Cardiol. 2012, 28, 288–295. [Google Scholar] [CrossRef]
  124. Miller, A.A.; Drummond, G.R.; Sobey, C.G. Reactive oxygen species in the cerebral circulation: Are they all bad? Antioxid. Redox Signal 2006, 8, 1113–1120. [Google Scholar] [CrossRef]
  125. Förstermann, U.; Li, H. Therapeutic effect of enhancing endothelial nitric oxide synthase (eNOS) expression and preventing eNOS uncoupling. Br. J. Pharmacol. 2011, 164, 213–223. [Google Scholar] [CrossRef]
  126. Smith, W.L.; DeWitt, D.L.; Garavito, R.M. Cyclooxygenases: Structural, cellular, and molecular biology. Annu. Rev. Biochem. 2000, 69, 145–182. [Google Scholar] [CrossRef]
  127. Rouzer, C.A.; Marnett, L.J. Mechanism of free radical oxygenation of polyunsaturated fatty acids by cyclooxygenases. Chem. Rev. 2003, 103, 2239–2304. [Google Scholar] [CrossRef]
  128. Hasan, D.; Hashimoto, T.; Kung, D.; Macdonald, R.L.; Winn, H.R.; Heistad, D. Upregulation of cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) in wall of ruptured human cerebral aneurysms: Preliminary results. Stroke 2012, 43, 1964–1967. [Google Scholar] [CrossRef]
  129. Vignais, P.V. The superoxide-generating NADPH oxidase: Structural aspects and activation mechanism. Cell Mol. Life Sci. 2002, 59, 1428–1459. [Google Scholar] [CrossRef]
  130. Liu, X.; Qin, Z.; Liu, C.; Song, M.; Luo, X.; Zhao, H.; Qian, D.; Chen, J.; Huang, L. Nox4 and soluble epoxide hydrolase synergistically mediate homocysteine-induced inflammation in vascular smooth muscle cells. Vascul. Pharmacol. 2019, 120, 106544. [Google Scholar] [CrossRef]
  131. Tamura, T.; Jamous, M.A.; Kitazato, K.T.; Yagi, K.; Tada, Y.; Uno, M.; Nagahiro, S. Endothelial damage due to impaired nitric oxide bioavailability triggers cerebral aneurysm formation in female rats. J. Hypertens. 2009, 27, 1284–1292. [Google Scholar] [CrossRef] [PubMed]
  132. Brandes, R.P.; Kreuzer, J. Vascular NADPH oxidases: Molecular mechanisms of activation. Cardiovasc. Res. 2005, 65, 16–27. [Google Scholar] [CrossRef]
  133. Tan, X.; Li, T.; Zhu, S.; Zhong, W.; Li, F.; Wang, Y. Induction of SPARC on Oxidative Stress, Inflammatory Phenotype Transformation, and Apoptosis of Human Brain Smooth Muscle Cells Via TGF-β1-NOX4 Pathway. J. Mol. Neurosci. 2020, 70, 1728–1741. [Google Scholar] [CrossRef]
  134. Juvela, S.; Poussa, K.; Porras, M. Factors affecting formation and growth of intracranial aneurysms: A long-term follow-up study. Stroke 2001, 32, 485–491. [Google Scholar] [CrossRef] [PubMed]
  135. Arnal, J.F.; Dinh-Xuan, A.T.; Pueyo, M.; Darblade, B.; Rami, J. Endothelium-derived nitric oxide and vascular physiology and pathology. Cell Mol. Life Sci. 1999, 55, 1078–1087. [Google Scholar] [CrossRef] [PubMed]
  136. Fukuda, S.; Hashimoto, N.; Naritomi, H.; Nagata, I.; Nozaki, K.; Kondo, S.; Kurino, M.; Kikuchi, H. Prevention of rat cerebral aneurysm formation by inhibition of nitric oxide synthase. Circulation 2000, 101, 2532–2538. [Google Scholar] [CrossRef]
  137. Förstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837, 837a–837d. [Google Scholar] [CrossRef]
  138. Sheinberg, D.L.; McCarthy, D.J.; Elwardany, O.; Bryant, J.P.; Luther, E.; Chen, S.H.; Thompson, J.W.; Starke, R.M. Endothelial dysfunction in cerebral aneurysms. Neurosurg. Focus. 2019, 47, E3. [Google Scholar] [CrossRef]
  139. Khurana, V.G.; Sohni, Y.R.; Mangrum, W.I.; McClelland, R.L.; O’Kane, D.J.; Meyer, F.B.; Meissner, I. Endothelial nitric oxide synthase gene polymorphisms predict susceptibility to aneurysmal subarachnoid hemorrhage and cerebral vasospasm. J. Cereb. Blood Flow. Metab. 2004, 24, 291–297. [Google Scholar] [CrossRef]
  140. Khurana, V.G.; Sohni, Y.R.; Mangrum, W.I.; McClelland, R.L.; O’Kane, D.J.; Meyer, F.B.; Meissner, I. Endothelial nitric oxide synthase T-786C single nucleotide polymorphism: A putative genetic marker differentiating small versus large ruptured intracranial aneurysms. Stroke 2003, 34, 2555–2559. [Google Scholar] [CrossRef]
  141. Sadamasa, N.; Nozaki, K.; Hashimoto, N. Disruption of gene for inducible nitric oxide synthase reduces progression of cerebral aneurysms. Stroke 2003, 34, 2980–2984. [Google Scholar] [CrossRef] [PubMed]
  142. Poole, J.J.A.; Mostaço-Guidolin, L.B. Optical Microscopy and the Extracellular Matrix Structure: A Review. Cells 2021, 10, 1760. [Google Scholar] [CrossRef] [PubMed]
  143. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef]
  144. Heino, J. The collagen family members as cell adhesion proteins. Bioessays 2007, 29, 1001–1010. [Google Scholar] [CrossRef]
  145. Klein, T.; Bischoff, R. Physiology and pathophysiology of matrix metalloproteases. Amino Acids 2011, 41, 271–290. [Google Scholar] [CrossRef]
  146. Chyatte, D.; Bruno, G.; Desai, S.; Todor, D.R. Inflammation and intracranial aneurysms. Neurosurgery 1999, 45, 1137–1146; discussion 1146–1137. [Google Scholar] [CrossRef]
  147. Aoki, T.; Kataoka, H.; Ishibashi, R.; Nozaki, K.; Hashimoto, N. Cathepsin B, K, and S are expressed in cerebral aneurysms and promote the progression of cerebral aneurysms. Stroke 2008, 39, 2603–2610. [Google Scholar] [CrossRef]
  148. Lepucki, A.; Orlińska, K.; Mielczarek-Palacz, A.; Kabut, J.; Olczyk, P.; Komosińska-Vassev, K. The Role of Extracellular Matrix Proteins in Breast Cancer. J. Clin. Med. 2022, 11, 1250. [Google Scholar] [CrossRef]
  149. Watton, P.N.; Selimovic, A.; Raberger, N.B.; Huang, P.; Holzapfel, G.A.; Ventikos, Y. Modelling evolution and the evolving mechanical environment of saccular cerebral aneurysms. Biomech. Model. Mechanobiol. 2011, 10, 109–132. [Google Scholar] [CrossRef]
  150. Nakagawa, D.; Zanaty, M.; Hudson, J.; Teferi, N.; Ishii, D.; Allan, L.; Jabbour, P.; Ortega-Gutierrez, S.; Samaniego, E.A.; Hasan, D.M. Plasma Soluble Human Elastin Fragments as an Intra-Aneurysmal Localized Biomarker for Ruptured Intracranial Aneurysm. J. Am. Heart Assoc. 2018, 7, e010051. [Google Scholar] [CrossRef]
  151. Yang, S.; Wang, T.; You, C.; Liu, W.; Zhao, K.; Sun, H.; Mao, B.; Li, X.; Xiao, A.; Mao, X.; et al. Association of polymorphisms in the elastin gene with sporadic ruptured intracranial aneurysms and unruptured intracranial aneurysms in Chinese patients. Int. J. Neurosci. 2013, 123, 454–458. [Google Scholar] [CrossRef] [PubMed]
  152. Onda, H.; Kasuya, H.; Yoneyama, T.; Takakura, K.; Hori, T.; Takeda, J.; Nakajima, T.; Inoue, I. Genomewide-linkage and haplotype-association studies map intracranial aneurysm to chromosome 7q11. Am. J. Hum. Genet. 2001, 69, 804–819. [Google Scholar] [CrossRef] [PubMed]
  153. Zhang, X.; Ares, W.J.; Taussky, P.; Ducruet, A.F.; Grandhi, R. Role of matrix metalloproteinases in the pathogenesis of intracranial aneurysms. Neurosurg. Focus. 2019, 47, E4. [Google Scholar] [CrossRef] [PubMed]
  154. Gross, J.; Lapiere, C.M. Collagenolytic activity in amphibian tissues: A tissue culture assay. Proc. Natl. Acad. Sci. USA 1962, 48, 1014–1022. [Google Scholar] [CrossRef]
  155. Hu, Z.; Deng, X.; Zhou, S.; Zhou, C.; Shen, M.; Gao, X.; Huang, Y. Pathogenic mechanisms and therapeutic implications of extracellular matrix remodelling in cerebral vasospasm. Fluids Barriers CNS 2023, 20, 81. [Google Scholar] [CrossRef]
  156. Tao, Z.; Jie, Y.; Mingru, Z.; Changping, G.; Fan, Y.; Haifeng, W.; Yuelan, W. The Elk1/MMP-9 axis regulates E-cadherin and occludin in ventilator-induced lung injury. Respir. Res. 2021, 22, 233. [Google Scholar] [CrossRef]
  157. Zhao, X.; Chen, J.; Sun, H.; Zhang, Y.; Zou, D. New insights into fibrosis from the ECM degradation perspective: The macrophage-MMP-ECM interaction. Cell Biosci. 2022, 12, 117. [Google Scholar] [CrossRef]
  158. Liu, P.; Shi, Y.; Fan, Z.; Zhou, Y.; Song, Y.; Liu, Y.; Yu, G.; An, Q.; Zhu, W. Inflammatory Smooth Muscle Cells Induce Endothelial Cell Alterations to Influence Cerebral Aneurysm Progression via Regulation of Integrin and VEGF Expression. Cell Transplant. 2019, 28, 713–722. [Google Scholar] [CrossRef]
  159. Kim, B.J.; Hong, E.P.; Youn, D.H.; Jeon, J.P. Genome-Wide Association Study of the Relationship Between Matrix Metalloproteinases and Intracranial Aneurysms. J. Clin. Neurol. 2022, 18, 163–170. [Google Scholar] [CrossRef]
  160. Aoki, T.; Kataoka, H.; Morimoto, M.; Nozaki, K.; Hashimoto, N. Macrophage-derived matrix metalloproteinase-2 and -9 promote the progression of cerebral aneurysms in rats. Stroke 2007, 38, 162–169. [Google Scholar] [CrossRef]
Figure 1. Structural changes during the formation of intracranial aneurysms. Under physiological conditions, the structure of cerebral arteries can be divided into three layers (from the inside out): (A) The intima, including the basement membrane (BM) and endothelial cells (ECs). (B) The middle membrane is composed of smooth muscle cells (SMCs) surrounding the direction of vessel, embedded with tightly arranged collagen and elastic fiber networks, ensuring good vascular compliance. (C) The outer membrane, mainly composed of collagen, maintains the integrity of the vascular wall structure and contains fibroblasts (FBs) and white blood cells (WBCs) [8,9]. There is an inner elastic layer (IEL) with elastic fibers between the intima and media, which is considered a key structure leading to the formation of aneurysms. The anatomical variations, structural composition, and physiological homeostasis of the cerebral arterial wall may be influenced by risk factors for intracranial aneurysm development, all of which contribute to abnormal blood flow. In response to these risk factors, structural changes in the cerebral arterial wall result in the disruption of the IEL at arterial bifurcations. Abnormal blood flow causes mechanical overload and tension transfer, leading to the continuous reconstruction and degradation of the extracellular matrix through SMC apoptosis or regulation as well as endothelial cell dysfunction and macrophage influx [10,11]. Blood inflow and impingement expose structurally deficient arteries to high wall shear stress, leading to the formation of an aneurysmal sac. The aneurysmal sac continues to grow until a balance is reached between continuous vascular wall repair and extracellular matrix degradation [12]. The main cellular components in the aneurysm wall are SMCs, a discontinuous layer of ECs, and a small number of inflammatory cells such as neutrophils, macrophages, and lymphocytes [9,13].
Figure 1. Structural changes during the formation of intracranial aneurysms. Under physiological conditions, the structure of cerebral arteries can be divided into three layers (from the inside out): (A) The intima, including the basement membrane (BM) and endothelial cells (ECs). (B) The middle membrane is composed of smooth muscle cells (SMCs) surrounding the direction of vessel, embedded with tightly arranged collagen and elastic fiber networks, ensuring good vascular compliance. (C) The outer membrane, mainly composed of collagen, maintains the integrity of the vascular wall structure and contains fibroblasts (FBs) and white blood cells (WBCs) [8,9]. There is an inner elastic layer (IEL) with elastic fibers between the intima and media, which is considered a key structure leading to the formation of aneurysms. The anatomical variations, structural composition, and physiological homeostasis of the cerebral arterial wall may be influenced by risk factors for intracranial aneurysm development, all of which contribute to abnormal blood flow. In response to these risk factors, structural changes in the cerebral arterial wall result in the disruption of the IEL at arterial bifurcations. Abnormal blood flow causes mechanical overload and tension transfer, leading to the continuous reconstruction and degradation of the extracellular matrix through SMC apoptosis or regulation as well as endothelial cell dysfunction and macrophage influx [10,11]. Blood inflow and impingement expose structurally deficient arteries to high wall shear stress, leading to the formation of an aneurysmal sac. The aneurysmal sac continues to grow until a balance is reached between continuous vascular wall repair and extracellular matrix degradation [12]. The main cellular components in the aneurysm wall are SMCs, a discontinuous layer of ECs, and a small number of inflammatory cells such as neutrophils, macrophages, and lymphocytes [9,13].
Ijms 26 03316 g001
Figure 2. Schematic diagram of potential sources and measurable types of biomarkers for intracranial aneurysms [27,28,29]. Cerebrovascular tissue, blood, and cerebrospinal fluid are potential sources of biomarkers [30,31,32].
Figure 2. Schematic diagram of potential sources and measurable types of biomarkers for intracranial aneurysms [27,28,29]. Cerebrovascular tissue, blood, and cerebrospinal fluid are potential sources of biomarkers [30,31,32].
Ijms 26 03316 g002
Figure 3. MiRNAs upregulated and downregulated during the formation of intracranial aneurysms [25,49,50,51,52,55,56,58,60,61,63,64,65,66,68,69,70,71,72].
Figure 3. MiRNAs upregulated and downregulated during the formation of intracranial aneurysms [25,49,50,51,52,55,56,58,60,61,63,64,65,66,68,69,70,71,72].
Ijms 26 03316 g003
Figure 4. Potential sources and roles of oxidative stress in the pathogenesis of intracranial aneurysms [118,119,120,122].
Figure 4. Potential sources and roles of oxidative stress in the pathogenesis of intracranial aneurysms [118,119,120,122].
Ijms 26 03316 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, E.; Yan, X.; Shen, H.; Zhao, M.; Gao, X.; Huang, Y. Intracranial Aneurysm Biomarkers: A Convergence of Genetics, Inflammation, Oxidative Stress, and the Extracellular Matrix. Int. J. Mol. Sci. 2025, 26, 3316. https://doi.org/10.3390/ijms26073316

AMA Style

Zhang E, Yan X, Shen H, Zhao M, Gao X, Huang Y. Intracranial Aneurysm Biomarkers: A Convergence of Genetics, Inflammation, Oxidative Stress, and the Extracellular Matrix. International Journal of Molecular Sciences. 2025; 26(7):3316. https://doi.org/10.3390/ijms26073316

Chicago/Turabian Style

Zhang, Enhao, Xu Yan, Hangyu Shen, Mingyue Zhao, Xiang Gao, and Yi Huang. 2025. "Intracranial Aneurysm Biomarkers: A Convergence of Genetics, Inflammation, Oxidative Stress, and the Extracellular Matrix" International Journal of Molecular Sciences 26, no. 7: 3316. https://doi.org/10.3390/ijms26073316

APA Style

Zhang, E., Yan, X., Shen, H., Zhao, M., Gao, X., & Huang, Y. (2025). Intracranial Aneurysm Biomarkers: A Convergence of Genetics, Inflammation, Oxidative Stress, and the Extracellular Matrix. International Journal of Molecular Sciences, 26(7), 3316. https://doi.org/10.3390/ijms26073316

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop